AU2005201938A1 - Amino acid sequences facilitating penetration of a substance of interest into cells and/or cell nuclei - Google Patents

Amino acid sequences facilitating penetration of a substance of interest into cells and/or cell nuclei Download PDF

Info

Publication number
AU2005201938A1
AU2005201938A1 AU2005201938A AU2005201938A AU2005201938A1 AU 2005201938 A1 AU2005201938 A1 AU 2005201938A1 AU 2005201938 A AU2005201938 A AU 2005201938A AU 2005201938 A AU2005201938 A AU 2005201938A AU 2005201938 A1 AU2005201938 A1 AU 2005201938A1
Authority
AU
Australia
Prior art keywords
lys
arg
amino acid
gly
leu
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2005201938A
Inventor
Eustrate Avrameas
Therese Ternynck
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Diatos SA
Original Assignee
Diatos SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU40748/01A external-priority patent/AU780785B2/en
Application filed by Diatos SA filed Critical Diatos SA
Publication of AU2005201938A1 publication Critical patent/AU2005201938A1/en
Abandoned legal-status Critical Current

Links

Description

AUSTRALIA
Patents Act 1990 COMPLETE SPECIFICATION STANDARD PATENT Applicant: DIATOS S.A.
Invention Title: AMINO ACID SEQUENCES FACILITATING PENETRATION OF A SUBSTANCE OF INTEREST INTO CELLS AND/OR CELL NUCLEI The following statement is a full description of this invention, including the best method of performing it known to us: SAMINO ACID SEQUENCES FACILITATING PENETRATION OF A SUBSTANCE OF INTEREST INTO CELLS AND/OR CELL NUCLEI The subject of this invention is an amino acid sequence having the capacity to facilitate k\ penetration of a substance of interest into cells and/or cell nuclei.
Having tools that make it possible to transfer substances of interest effectively from the outside medium to the inside of cells, and more specifically cell nuclei, is a major advantage in the field of C biotechnology, especially to produce proteins or peptides to regulate the expression of genes, or to analyze 0\ and screen intracellular signaling paths, or to analyze the properties of a given substance on said cell.
Cl Another important application of such tools concerns the field of gene therapy since, up until the present time, the various gene therapy processes have run into the same need, not met in an optimal way, Cxl which is to be able to have vectors that can transfer biologically active ingredients into the cytoplasm and/or the cell nucleus from the host organism being treated, without thereby altering either the genome of the host or the biological properties of said active ingredients transferred.
Several techniques have been developed thus far to transfer DNA into cells, but none is really satisfactory. One of them, derived from the Mandel and Higa technique, based on acquisition by E. coli of susceptibility to transformation by treatment with CaCI 2 consists of co-precipitating DNA with calcium phosphate or DEA dextran, and introducing the precipitate obtained directly into the cell or nucleus. This method, besides the fact that it is toxic, is not selective.
Another method of directly introducing DNA, electroporation, consists of giving the cells a brief electric shock of several thousand volts, which allows the DNA to pass through the cytoplasmic membrane and to be introduced into the cell. This method is very toxic to the cells, involving high mortality and great variability, depending on the cells used.
Other methods use receptors on the membrane to screen the entry of the gene into the cells. The DNA can then penetrate into the cell through either a specific ligand of these receptors, or specific antibodies of membrane constituents. The DNA-ligand complex thus penetrates into the cell by a process of endocytosis. This process is limited due to the fact that there is major destruction of the complex used in the lysosomal vesicles. Several processes have been developed to eliminate these disadvantages, but none gives complete satisfaction.
US patent No. 5 635 383 describes another type ofpolylysine-based complex vector for transferring nucleic acids into cells.
US patent No. 5 521 291 describes another method based on the use of a conjugate formed from a virus linked to a substance having a strong affinity for DNA via an antibody. Such conjugates are heavy to use, and certain risks are associated with the use of viruses.
tl To try to eliminate these disadvantages, a process was described in patent application No. WO O 97/02840 that is used in vitro and consists of using anti-DNA murine antibodies, or their fragments F(ab')2 and Fab', capable of penetrating inside live cells, as immunovectors for intracytoplasmic and/or C intranuclear transfer of biologically active substances. Although these vectors are highly effective, they can be complicated to use in some applications. Using molecules the size and complexity of antibodies can also
N
kO be a major disadvantage in their handling and implementation.
Patent application No. WO 99/07414 described how it was possible to use in vitro peptides 00 derived from anti-DNA murine antibodies, divulged in the above-mentioned application WO 97/02840, as Sintracytoplasmic and intranuclear internalization vectors for biologically active substances.
SAlthough these murine-origin peptide vectors are coded by the germinal line and carry no lt mutation, and consequently, should be antigenically close to those encountered in humans, the risk of an immune reaction in humans cannot be excluded.
(N
There is therefore a real need for peptides and amino acid sequences that eliminate the disadvantages described above, namely that can be used as, or in, a cell internalization vector in humans and would pose none of the risks mentioned above.
,t is known that a very large number of cell regulations depend on interactions between the proteins and the glycosaminoglycans (GAG) on the surface of cells [the numbers in bold, in parentheses, refer to the list of bibliographic references attached]. Such interactions occur, for example, in the control of hemostasis in the proliferation of smooth muscle cells in the expression of the activity of the growth factors in the expression of the lipolytic activity of enzymes in the integrity of the extracellular matrix (11) and others. In a given system, these biological effects are due to the interaction of one or a limited number of proteins with the GAG on the cell surface. The GAG are constituents very well preserved in the course of evolution, present on the surface of all eukaryotic cells.
The GAG are saccharide polymers; for example, heparin is a polymer of disaccharide 4-2-iduronic acid D-glucosamine and the chondroitine sulfates are polymers of disaccharide N-acetyl chondrosine, containing a very large number of sulfate groups and hence negatively charged. The proteins that react specifically with the GAG all contain, in their sequence, one or more peptide segments that are responsible for the interaction of those proteins with the GAG. The length of these segments varies from one protein to another, ranging from four to thirty amino acids. Nearly all these peptides are positively charged and contain a high number of basic amino acids, above all lysine and arginine. It has been shown that these amino acids participate in preponderant fashion in the interaction of these proteins with the GAG The peptides binding to the GAG or, more generally, to the aminoglycans, and in particular to heparin, heparin sulfate and the chondroitine sulfates (peptides generally designated by HBP, for "heparinbinding peptide," even if heparin is only one example of aminoglycan) can be natural in origin, like the peptides described above, or artificial. They can be used in their natural or polymer (dimer, trimer, etc.) O form.
It has been found, unexpectedly, in this invention, that these peptides can be used both in vivo and in vitro as agents for internalizing substances of interest into cells.
IN Indeed, using a known in vitro technique in vivo involves determining several parameters (bioavailability, immune reactions, etc.), which do not occur during in vitro tests, in a closed space. In addition, such a transfer from in vitro to in vivo remains rather hazardous, in sum, due to the many potential 00 C interactions that can interfere with manipulation in vivo (immune reaction, side effects, blockage or inhibition of active ingredients, rejection, etc.); consequently, there is little hope of reasonable success Susing an in vitro technique in vive.
Thus, according to a first aspect, the subject of this invention is an amino acid sequence that has the capacity to facilitate penetration of a substance of interest inside cells and/or cell nuclei and that is characterized by the fact that it is capable of reacting in vivo with the aminoglycans.
According to another aspect, the subject of this invention is an amino acid sequence that has the capacity to facilitate penetration of a substance of interest inside cells and/or cell nuclei and is characterized by the fact that it comes from a protein of human origin and is capable of reacting with the aminoglycans.
More specifically, in one case or another, this sequence is characterized by the fact that it reacts with heparin, the chondroitine sulfates and their derivatives.
The research work done as part of this invention has made it possible to show that penetration of the peptides is completely inhibited by incubating the cells at 4 0 C. It. is also partly inhibited by inhibitors of cell metabolism, like sodium azide (inhibitor of ATPase), genistein (inhibitor oftyrosine kinase and the bond to ATP). The mechanism for internalization of the peptides in the invention, and hence of the substances of interest coupled to said peptides, is therefore dependent on energy. The vectorization using the peptides in the invention is therefore remarkable by the fact that it does not come from a passive system but, on the contrary, is done via a receptor. The amino acid sequences in the invention are therefore characterized, besides their capacity to react in vivo with the aminoglycans, the aminoglycan sulfates, the chondroitines and the chondroitine sulfates, by their capacity to be fixed to a receptor of the cell membrane and to cross said cell membrane thanks to that receptor. Thus, the amino acid sequences in the invention are distinguished from the peptide transporters in the prior art that were capable of crossing the cell membrane in a passive way.
The peptides in the invention are therefore remarkable in that they have the capacity to be able to cross the cell membranes by an active mechanism, then to lodge in the cytoplasm and/or the cell nucleus and thus to make it possible to have a vector whose use is not limited, when it passes into the cell, by the size of the substances being transported. Indeed, the vectors in the invention are capable of transporting drugs, ranging from small chemical molecules (low molecular weight) to proteins or plasmid-type nucleic acids (high molecular weight). The use of these vectors thus opens a new path in intracellular protein therapy or gene therapy. This special capacity of the vectors in the invention for penetration makes it possible to target "drugs" in the cells in a preferential way, thus contributing to a potential reduction in the toxicity of the drugs and a potential increase in the efficacy index.
"Heparin or chondroitine sulfate derivatives" or "aminoglycans like heparin or chondroitine sulfate" are understood to mean any product or sub-product as defined in the publications cited in references to 00 00"Facilitate penetration" is understood to mean facilitating the passage or translocation of a substance from the outside medium into the intracellular medium, and quite specifically into the cytoplasm and/or the nucleus of the cell. This penetration can be determined by various processes, for example by a V)cell penetration test having a first incubation step for the amino acid sequence in the presence of culture cells, followed by a fixing step and permeabilization of those cells, and then revelation of the presence of C' said amino acid sequence inside the cell. The revelation step can be done with another incubation in the presence of antibodies marked and directed against said sequence, followed by detection in the cytoplasm or in immediate proximity of the cell nucleus, or even within it, of the immunologic reaction between the sequence and the marked antibodies. Revelation can also be done by marking an amino acid sequence in the invention and detecting the presence of said marking in the cell compartments. A cell penetration test was described in the above-mentioned patent application No. WO 97/02840.
"Substance of interest" is understood to mean any product presenting an interest, particularly biological, pharmaceutical, diagnostic, tracing, or agro-food. They can be nucleic acids (ribonucleic acid, deoxyribonucleic acid) that can have various origins, and particularly, human, viral, animal, eukaryotic or prokaryotic, vegetable, synthetic, etc., and able to vary in size, ranging from a simple oligonucleotide to a genome or genome fragment. It can also be a viral genome or plasmid. The substance can also be a protein, such as an enzyme, a hormone, a cytokine, an apolipoprotein, a growth factor, an antigen, an antibody, etc.
It can also be a toxin, antibiotic, antiviral molecule or immune modulator.
Generally, the substance of interest can be any active ingredient of a drug, whether it be a chemical, biochemical, natural or synthetic product. The molecules can be small, with a molecular weight of around 500 D, or large like proteins of several thousand Daltons.
The substance of interest can be directly active or can be activated in situ by the amino acid sequence, by a distinct agent or by environmental conditions.
The scope of the invention extends to combinations of the amino acid sequence with a substance of interest as defined above.
In one preferred embodiment of this invention, to eliminate the risks described above, the amino acid sequence is coded by the germinal line, and thus does not carry any mutations (substitution, deletion, addition, etc.).
SIn this invention, one especially preferred amino acid sequence comes from a protein synthesized by a human cell. Advantageously, said protein synthesized by a human cell is chosen from among the proteins linked to heparin-type or chondroitine sulfate-type aminoglycans. For example, it is an amino acid sequence that comes from human lipoprotein B.
Generally, the amino acid sequence includes a high number of basic amino acids, as is the case in Slysine, arginine or histidine, for example.
"High number" should be understood as at least equal to 3.
C One preferred type of amino acid sequence for use in this invention is composed of, or contains, at least one group of amino acids with one of the following formulas: a) (XBBBXXBX)n; b) (XBBXBX)n; c) (BBXmYBBXo)n d) (XBBXXBX)n; and e) (BXBB)n; in which: B is a basic amino acid; X is a non-basic, preferably hydrophobic amino acid, such as alanine, isoleucine, leucine, methionine, phenylalanine, tryptophan, valine or tyrosine; Y represents either a direct bond, or 1 to 20 amino acids; m is a whole number between 0 and 5; n is a whole number between 1 and preferably between 1 and 3; and o is a whole number between 0 and Generally, the amino acid sequences have less than 100 amino acids, less than 50 amino acids is considered better, and less than 25 amino acids better yet.
Advantageously, the amino acid sequences in the invention have between 6 and 25 amino acids.
Preferred amino acid sequences for use in this invention are those identified by SEQ ID NO: 2, 3, 7, 9, 10, 13, 16, 17, 19, 20, 21, 23, 25, 26, 30, 33, 34, 35, 36, 37,38 and 39 on the attached list, which is part of this description. Among them, those especially preferred are the sequences identified by SEQ ID NO: 2, 3, 5, 7, 9, 10, 13, 16, 19, 20, 21, 23, 25, 26, 30, 36 and 38.
Another preferred type of sequences, for use in this invention, is composed of or includes at least two domains, one of those domains containing an amino acid sequence with the formula: a) XBBBXXBX; b) XBBXBX; c) BBXmYBBXo; d) XBBXXBX; or e) BXBB; and the other of those domains having an amino acid sequence with the formula: a) XBBBXXBX; b) XBBXBX; c) BBXmYBBXo; d) XBBXXBX; 3) BXBB; or f) an antibody fragment; formulas in which: B is a basic amino acid, X is a non-basic amino acid, preferably hydrophobic, Y is either a direct bond, or 1 to 20 amino acids, m is a whole number between 0 and 5, o is a whole number between 0 and One particularly interesting amino acid sequence is the sequence SEQ ID NO: 1, to the extent that in the state at least of dimer, it has the properties expected and in the state of monomer or polymer, it gives another amino acid sequence to which it is coupled said properties or substantially potentiates those properties when said sequence already has them. Similarly, the peptides designated HBP3, HBP7, (HBP3)2, HBP6, HBP7, HBP10 and HBP13 have this potentiation capacity.
The peptides as defined above are capable of transporting inside cells molecules that are combined with them covalently or non-covalently, and are thus effective vectors for intracellular transfer of the substances of interest.
The seven sequences described in Table 1 below are particularly useful for intracytoplasmic transport. They are sequences (HB1)3, HBP6, (HBP3)2, HBP7, HBP11, HBP 13 and HBP2.
Table 1 %K %R Identity Sequence AA Total residues Total residues K R (HB1)3 VKRGLKLVKRGLKRVKRGLKR 28 24 52 HBP6 GRPRESGKKRKRKRKRLKP 32 31 63 (HBP)2 SERKKRRRESRKKRRRES 22 44 67 HBP7 GKRKKKGKLGKKRDP 46 13 HBP11 AKTGKRKRSG 30 20 HBP13 KHLKKHLKKHLK 50 0 HBP2 RKGKFYKRKQCKPSRGRK 33 22 Two sequences are particularly useful for intranuclear transport. They are sequences HBP10 and These two sequences are characterized by their high basic amino acid content, and more particularly by the fact that they do not contain any lysine residue, contrary to the sequences useful for intracytoplasmic transport.
Each of the sequences described in Table 1 has at least 20% lysine residues, and at least 50% of the residues are basic amino acids out of the total number of residues in the sequence.
This invention shows that coupling peptides, as defined above, with polypeptide vectors capable of penetrating inside cells substantially increases the translocation power of those vectors.
This invention also shows that coupling peptides, as defined above, or their polymer forms, with a ligand, whose function is to react with a receptor present on the cell membrane, substantially increases that ligand's capacity to attach itself to the cell membrane.
In another of its aspects, this invention is aimed at using the amino acid sequences defined above to prepare compositions designed to transfer substances of interest into cells. This capacity of the peptides in the invention is an advantage to allow the transport of active substances through biological membranes and, quite specifically, through the hematoencephalic, hematoretinal, intestinal and pulmonary barriers. The peptides in the invention have the interest of being able to be used in forms of administration adjusted to both the active substance to which they are coupled and to the type of cell targeted, particularly those requiring passage through the above barriers.
In another embodiment, this invention concerns the use of said amino acid sequences in a peptide vector. These vectors, due to the properties of said amino acid sequences, can be used easily for intracytoplasmic and intranuclear transfer in humans, with no risk to them or any degradation of the substance of interest coupled to the vector.
To achieve this goal, it is necessary for a vector to be capable of transporting relatively large quantities of molecules inside cells and for it not to be recognized as a foreign antigen by the human immune system.
Table 1 below gives, for several peptides in the invention, their level of intracytoplasmic and/or intranuclear penetration coupled to a low-molecular weight marker allowing their detection as biotin or fluorescein (Marq) or to a biological substance of interest (Subst).
The results given in Table 1bis are based on the experimental data reported hereinafter.
Table Ibis Peptide SEQ ID NO Intracytoplasmic Intranuclear Marq Subst Marq Subst HBP1 1 ND (HBP1) 2 2 (HBP1) 3 3 4 6 7 8 9 11 12 13 14 16 HBP4 17 ND 18 ND 2 19 HBP2 20 HBP6 21 22 23 24 (HBP3) 2 26 27 28 29 HBP7 30 31 32 HBP8 33 HBP9 34 HBPIO 35 HBPll 36 HBP12 37 HBP13 38 39 These results indicate that the peptides that are capable of massive nuclear translocation are those whose positive charge is mainly comprised of the presence of arginine and the almost total absence of lysine.
The first group of peptides in the invention includes amino acid sequences capable of allowing penetration of a substance of interest inside the cell, but little or none inside the nucleus. Examples that can be cited are the peptides in sequences SEQ ID NO: 2, 3, 17, 18, 19, 20, 21, 33, 34, 37 and 38.
The second group of peptides in the invention has amino acid sequences capable of allowing penetration of a substance of interest inside the cell and the cell nucleus. Examples that can be cited are the peptides in sequences SEQ ID NO: 26, 35 and 39.
A vector in this invention is characterized by the fact that it is composed of, or includes, an amino acid sequence as defined above.
As a variation, the vector is based on coupling, on one hand, of amino acid sequences reacting with aminoglycans and, on the other hand, of new peptidesderived from the variable part of human anti- DNA antibodies. The coupling of amino acid sequences reacting with aminoglycans and peptides derived from variable parts of human anti-DNA antibodies inside one and the same molecule results in the preparation of a peptide vector that is particularly effective in translocation and intracellular transfer of O substances of interest, above all when the amino acid sequences reacting with the aminoglycans are human in origin.
This combination also gives rise to a translocation and transfer vector specially adapted for use in NO humans. Indeed, as indicated above, although the peptide vectors ofmurine origin known from WO O 97/02840 are coded by the germinal line and carry no mutations, and consequently should be close to those encountered in humans in terms of antigens, it is possible that their injection into humans would induce an 00 immune reaction. The peptide vector formed from HBP according to this invention and from peptides Sderived from anti-DNA antibodies, both of human origin, coded by the germinal line and carrying no mutations, prevents this problem.
In The general characteristics of these peptides derived from human anti-DNA antibodies are close to those of the peptides of murine origin described in patent application WO 99/07414, while they have additional properties that distinguish them from the latter, namely: 1) To penetrate inside cells, they have to have an active cell metabolism (culture temperature between 25°C and 39°C, and preferably 37 0 while the murine peptides are clearly less dependent; 2) They react much less strongly with DNA than the murine vectors; 3) Their penetration capacity is not significantly influenced by the molecule they are going to transport inside the cell; 4) They penetrate better inside cells of human origin than inside cells of other origins.
This invention provides a vector composed ofa HBP and one or more antibody fragments, preferably polyreactive, and more specifically one or more fragments that come from hypervariable regions of the antibody. Preferably, the vector that is the subject of the invention is characterized by the fact that it contains a fragment of the heavy chain of an antibody.
In the above-mentioned patent application WO 99/07414, only fragments of a monoclonal IgG were used, which is a monomer immunoglobulin that is small in size and has a low molecular weight. This invention shows, for the first time, that it is also possible to use a fragment that comes from an IgM, which is a pentamer immunoglobulin, with a very high molecular weight. Indeed, until now, to the knowledge of the applicant, no one has done any research on using an IgM fragment as a vector for cell internalization, due to the dissuasive character of the large size and high molecular weight of such fragments.
The subject of this invention is therefore a vector for cell internalization, characterized by the fact that it contains one or more HBP and one or more fragments of an IgM or an IgG.
I
Preferably, said vector contains all or part of the CDR2 region of an antibody. As a variation, said vector contains all or part of the CDR3 region of an antibody. More specifically, said vector contains at least one CDR3 region of an anti-DNA human antibody, selected from the group consisting of RTT79, NE- S and RT72.
In another variation, the vector that is the subject of this invention can also contain all or part of region CDR2, and all or part of region CDR3.
By "all or part" it is understood that the vector in the invention can contain either the whole CDR 00 Sregion concerned, or only part of it, provided that the vector retains the capacity to penetrate into the cells (functional homologue). By "part of CDR region" is understood a CDR region deprived of one or more terminal amino acids. It can also be a CDR region in which one or more internal residues have been deleted or substituted for other amino acids, preferably amino acids of the same nature (basic amino acids, for example).
Some of the examples given in this patent application are based on the use of SEQ ID NO: 1 which comes from human lipoprotein B, but for a person skilled in the art, it is obvious that any natural or artificial HBP can be used.
As indicated above, the vector in the invention is particularly well suited for intracellular and intranuclear transport and transfer of substances of interest.
This invention is therefore aimed at supplying a vector such as the one described above, characterized by the fact that it contains a substance of interest naturally, or not naturally, that can be incorporated into the cells and/or the nuclei of said cells.
More specifically, the subject of this invention is a vector whose penetration capacity is quite independent from the nature of the substance of interest that is coupled to it. This characteristic, proper to these human vectors compared to the murine vectors, is of primary interest in the planned use of these vectors. But the invention is also interested in vectors that are adapted to the substance of interest which is coupled to it.
"Coupling" is understood to mean any type of interaction allowing a physical association between the substance of interest and the vector. It can be cleavable or non-cleavable according to the biological medium and/or the substance of interest transported by the peptides of the invention or it can be cleavable by physical means applied to the organism to which the vector coupled to the active substance has been administered. Thus, the expression of the biological effect of the substance can require that it be released from the vector. Doxorubicin can be cited as an example of a substance of interest that is preferably released from the vector.
However, the interaction must be solid enough that the vector does not dissociate before or during cell penetration. For this reason, the coupling preferred in the invention is covalent coupling, although it can be non-covalent coupling. The substance of interest can be coupled directly to the peptide either on one of those terminal ends or on a side chain or one of the amino acids. The substance of interest can also be O coupled indirectly by a connecting arm either to one of the terminal ends of the peptides or to a side chain of one of the amino acids.
The coupling can be done by any chemical, biochemical, enzymatic or genetic coupling process N known to a person skilled in the art, but it is generally preferred to use a homofunctional or Sheterofunctional bridging reagent like succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC). Other coupling means that can be cited are those chosen from among: bi-functional or multi- 00 functional agents containing alkyl, aryl, aralkyl or peptide groups; esters; aldehydes or alkyl, aryl or aralkyl 0\ acids; anhydride, sulfhydrile or carboxyl groups such as maleymil benzoic acid and maleymil propionic acid derivatives and succinimidyl derivatives; groups derived from bromide or cianogenic chloride, Scarbonyldiimidazole, succinimide esters or sulphonic halogenides.
In another form of embodiment of this invention, said substance of interest can also be coupled by any genetic engineering technique known to a person skilled in the art. "Genetic engineering" means using an expression vector in which the DNA coding for peptide vectors is cloned in phase in 5' and/or 3'of the complementary DNA of the gene of interest. Expression of the fusion protein is under the control of a promotor. The expression system can be used in a prokaryotic or eukaryotic host cell for the production of the fusion protein.
In a first form of embodiment, said substance of interest is coupled at the N-terminal end of the amino acid sequence in the invention. In a second form of embodiment of the invention, said substance of interest is coupled at the C-terminal end of said sequence.
Surprisingly, it has been shown that the vector that is the subject of the invention is capable of potentiating the biological activity and, potentially, reducing the toxicity of said coupled substance. This invention therefore also has as its subject a vector characterized by the fact that it makes it possible to increase the biological activity of the substance of interest to which it is coupled.
It has also been shown that the vector that is the subject of the invention permits transfection of cells in vitro.
In one particular embodiment of the invention, the vector is coupled to the substance of interest by at least one molecule (called an "anchoring molecule") that has a strong natural affinity for the substance of interest to be internalized. The natural affinity of the anchoring molecule for the substance of interest allows the transporter to interact non-covalently with said substance of interest, and hence to carry it along in intracellular travel.
Another especially interesting advantage of this type of transporter consists of the fact that, due to the natural affinity of the anchoring molecule for the substance of interest, these two elements are coupled in a totally natural way, with no chemical or biochemical interaction.
I
n This type of transporter is particularly interesting in a case where the substance of interest, due to its size and/or its structure, proves difficult to couple directly to the amino acid sequence. This type of transporter can also prove particularly useful when the substance of interest is not very stable, and when any kind of chemical interaction for coupling it could degrade it or alter its activity.
In addition, the transporter in the invention cannot be specific for a single substance of interest, but can, on the contrary, permit internalization of several different substances of interest inside cells and/or cell nuclei.
00
M
0 The invention also concerns eukaryotic cells that contain an amino acid sequence in the invention.
SIt also concerns eukaryotic cells that contain a vector and/or a transporter according to the invention. It also Sconcerns any type of eukaryotic cell that has been transfected by a vector and/or transporter according to t this invention.
The invention also concerns a process to transfer a substance of interest inside a cell in vitro and to increase the biological activity of said substances of interest that has the following steps: a) coupling the substance to an amino acid sequence, to a vector or to a transporter according to the invention, as described above, and b) incubating the cell with said coupling product at a culture temperature that permits active metabolism of said cell.
Such a temperature is between 25°C and 39 0 C, preferably 37'C.
This invention also concerns a composition having as its active ingredient either vectors or transporters loaded with at least one substance of interest according to this invention, or eukaryotic cells that have been transfected according to this invention. Its subject is also the use of such compositions for the formulation and preparation of biological, pharmaceutical, cosmetic and agro-food products.
The scope of the invention extends to pharmaceutically acceptable alkaline or acidic addition salts, hydrates, esters, solvates, precursors, metabolites or stereoisomers, of said vectors and transporters loaded with at least one substance of interest. The scope of the invention also extends to pharmaceutical formulations having a vector or transporter loaded with at least one substance of interest in combination with a pharmaceutically acceptable vehicle, diluent or excipient.
The expression "pharmaceutically acceptable salts" refers to non-toxic salts of the amino acid sequences in the invention that can generally be prepared by having the free base react with a suitable organic or inorganic acid. These salts retain their biological efficacy and the properties of the free bases.
Representative examples of said salts that can be cited are water-soluble and water-insoluble salts, such as acetates, ansonates (4,4-diaminostilbenes- 2 2 '-disulfonates), benzene sulfonates, benzonates, bicarbonates, bisulfates, bitartrates, borates, bromides, butyrates, calcium edetates, camsylates, carbonates, chlorides, citrates, clavulariates, dichlorohydrates, edetates, edisylates, estolates, esylates, fumarates, gluceptates, gluconates, glutamates, glycolylarsanylates, hexafluorophosphates, hexylresorcinates, hydrabamines, 0 hydrobromides, hydrochlorides, hydroxynaphthoates, iodides, isothionates, lactates, lactobionates, laurates, 1 malates, maleates, mandelates, mesylates, methyl bromides, methyl nitrates, methyl sulfates, mucates, napsylates, nitrates, 3-hydroxy-2-naphthoates, oleates, oxalates, palmitates, pamoates (1,l-methylene-bis-2hydroxy-3-naphtoates, emboates), pantothenates, phosphates/diphosphates, picrates, polygalacturonates, propionates, p-toluene sulfonates, salicylates, stearates, subacetates, succinates, sulfates, sulfosalicylates, Ssuramates, tannates, tartrates, teoclates, tosylates, triethiodides, valerates and salts of N-methylglucamine ammonium.
OO A subject can be treated with a pharmaceutically effective quantity of a peptide, a vector or a Stransporter according to the invention, loaded with at least one substance of interest. The expression "pharmaceutically effective quantity" means a quantity capable of making the substance of interest penetrate sufficiently to induce the biological or medical response of a tissue, system, animal or human as Sexpected by the researcher or attending physician.
C N The invention also covers pharmaceutical compositions suitable for the introduction of a substance of interest into a cell or cell nucleus. The compositions contain an effective quantity of a vector or transporter according to the invention, loaded with at least one substance of interest, alone or in combination with one or more pharmaceutically acceptable supports. The compositions are particularly useful in the sense that they have very low toxicity, or are non-toxic.
The vectors or transporters in the invention, or their salts, loaded with at least one substance of interest, can be administered by any of the routes of administration accepted for therapeutic agents. These processes include systemic administration, for example oral, nasal, parenteral or topical administration, for example transdermal or even central administration, for example by the intracranial surgical route or intraocular administration.
Oral administration can be used by means of tablets, gel capsules, soft capsules (including delayed or extended-release formulations), pills, powders, granules, elixirs, tinctures, suspensions, syrups and emulsions. This form of presentation is particularly suitable for passage through the intestinal barrier.
Parenteral administration is generally done by subcutaneous, intramuscular or intravenous injection or by perfusion. The injectable compositions can be prepared in the traditional forms, either in suspension or liquid solution or in solid form for extemporaneous dissolution in a liquid. This form of presentation is more specifically adapted for passage through the hematoencephalic barrier.
One possibility of parenteral administration uses implantation of a slow or extended-release system that makes sure a constant dosage level is maintained, for example according to US-A-3 710 795.
For intranasal administration, suitable intranasal vehicles can be used.
For transdermal administration, transdermal cutaneous stamps well known to a person skilled in the art can be used. A transdermal-release system permits continuous administration.
Other preferred topical preparations include creams, unguents, lotions, aerosol sprays and gels.
O Based on the planned route of administration, the compounds can come in solid, semi-solid or Sliquid form.
For the solid compositions, such as tablets, pills, powders or granules in the free state or included in gel caps, the active ingredient can be combined with: a) diluents, for example lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, for example silicone, talc, stearic acid, its Smagnesium or calcium salt and/or polyethylene glycol; c) binders, for example magnesium and aluminum silicate, starch paste, gelatin, tragacanth gum, methyl cellulose, sodium carboxymethyl cellulose and/or 00 polyvinyl pyrrolidone; if necessary, d) disintegrators, for example, starch, agar, alginic acid or its sodium S salt, or effervescent mixtures; and/or e) absorbents, dyes, aromatic agents and sweeteners. The excipients can be, for example, mannitol, lactose, starch, magnesium stearate, sodium saccharine, talc, cellulose, r, glucose, sucrose, magnesium carbonate and pharmaceutical quality analogs.
SFor semi-solid compositions, such as suppositories, the excipient can be, for example, an emulsion or fatty suspension, or one with a polyalkylene glycol base, such as polypropylene glycol.
The liquid compositions, particularly injectables or those to be included in a soft capsule, can be prepared, for example, by dissolution, dispersion, etc. of the active ingredient in a pharmaceutically pure solvent such as, for example, water, physiological serum, aqueous dextrose, glycerol, ethanol, an oil and the like.
The vectors or transporters in the invention, loaded with at least one substance of interest, can also be administered in the form of liposome-type release systems, such as in the form of small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. The liposomes can be made from a variety ofphospholipids, containing cholesterol, stearoylamine or phosphatidyl cholines. In one form of embodiment, a film of liquid components can be hydrated with an aqueous solution of the drug to form a lipid layer encapsulating the medication, as described in US-A-5 262 564.
The compounds in the invention can be sterilized and/or contain adjuvants and non-toxic auxiliary substances, such as preserving, stabilizing, wetting or emulsifying agents, agents promoting dissolution, salts to regulate osmotic pressure and/or buffers. They can also contain other substances that have therapeutic interest. The compositions are prepared, respectively, by traditional mixing, granulating or coating processes, and they contain around 0.1% to 75%, preferably around 1% to 50% active ingredient.
The vectors or transporters in the invention, loaded with at least one substance of interest, can also be coupled with soluble polymers, such as targetable drug supports. Such polymers can contain polyvinyl pyrrolidone, pyrane copolymer, polyhydroxypropyl-methacrylamide-phenol, polyhydroxy-ethylaspanamide-phenol or poly (ethylene oxide)-polylysine substituted with palmitol residues. The compounds in this invention can also be coupled to a class of biodegradable polymers useful in producing controlled release of a drug, for example, poly(lactic acid), poly(epsilon-caprolactone), poly(hyroxybutyric acid), the polyortho esters, the polyacetals, the polydihydropyranes, the polycyanoacrylates and the reticulated or amphipathic sequenced hydrogel copolymers.
The dosage for the administration of the vectors or transporters in the invention, loaded with at least one substance of interest, is chosen depending on a diversity of factors including the type, species, age, weight, sex and medical condition of the subject: the seriousness of the \Ocondition being treated; the route of administration; the status of the subject's kidney and liver functions, and the nature of the particular compound or salt used. A regularly experienced 00 physician or veterinarian will easily determine and prescribe the effective quantity of the vector or transporter loaded with the substance of interest in order to prevent, thwart or stop the progress of the medical condition being treated.
Any of the above pharmaceutical compositions can contain from 0.1% to 99%, preferably 1% to 70%, of the active ingredient.
SAs examples, the oral dosages of the vectors or transporters in the invention, loaded with at least one substance of interest, when they are used for the effects indicated, will be between around 0.05 and 1,000 mg/day by the oral route and, preferably come in the for of tablets containing 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100.0, 250.0, 500.0 and 1,000.0 mg of active ingredient. The effective plasma levels of the vectors or transporters loaded with at least one substance of interest will range from 0.002 mg to 50 mg per kilogram of body weight and per day.
The vectors or transporters in the invention, loaded with at least one substance of interest, may be administered in the form of single daily doses, or thetotal daily dose may be administered in two, three or four doses per day.
In one particular application, this invention relates to a diagnostic agent for in vitro use, composed of or containing at least one vector, one transporter and/or one cell according to the invention. Such a diagnostic agent can also be used in vivo.
The subject of this invention is therefore also a diagnostic kit that includes said diagnostic agent. More specifically, the diagnostic kit includes a predetermined quantity of a composition in the invention, in one or more containers.
Similarly, the amino acid sequence in the invention, or vector and/or a transporter containing that amino acid sequence, or cells transfected with the help of said vector, can be used in vivo for preventative purposes, for example and in a non-limiting way, for the prevention of viral infections, metastases, cell apoptosis (degenerative diseases, tissue ischemia), or for therapeutic purposes, for example the treatment of infectious diseases (viral, bacterial), cancer and pathological neo-angiogensis.
In the claims which follow and in the description of the invention, except where the context requires otherwise due to express language or necessary implication, the word "comprise" or variations such as "comprises" or "comprising" is used in an inclusive sense, i.e.
t to specify the presence of the stated features but not to preclude the presence or addition of N further features in various embodiments of the invention.
All references, including any patents or patent applications, cited in this specification are hereby incorporated by reference. No admission is made that any reference ND 5 constitutes prior art. The discussion of the references states what their authors assert, and the 0 applicants reserve the right to challenge the accuracy and pertinency of the cited documents. It will be clearly understood that, although a number of prior art publications are referred to 00 herein, this reference does not constitute an admission that any of these documents forms part of Sthe common general knowledge in the art, in Australia or in any other country.
Other advantages and characteristics of the invention will appear from the examples of t embodiment that follow and refer to the attached drawings.
16a I MATERIALS AND METHOD.
S1) Cell Lines.
a) Normal Cells: PtK2, kangaroo rat kidney fibroblasts 3T3, mouse embryo fibroblasts S- HUVEC, human endothelial cells CHO, Chinese hamster ovaries 0- HUVEC, endothelial cells transfected by a cell multiplication gene C CHO, hamster kidney cells S- CHO-745, cells from the CHO line defective for the synthesis of xylosyl transferase b) Tumor Cells: H1299, human pulmonary carcinoma HH9, tumor epithelial cells from the human breast transfected by a gene coding for a growth factor SMCF7, human tumor epithelial cells MCF7 ras, MCF7 cells transfected by the ras gene HeLa, carcinoma from the neck of the human uterus HCT 116, carcinoma from the human colon HT-29, adenocarcinoma from the human colon SLS 174T, adenocarcinoma from the human colon B16-F10, cells from murine melanoma Daudi, human Burkitt's lymphoma c) Cell Culture: The cells are cultivated in a DMEM medium containing 2% L-glutamine, 1% sodium pyruvate, penicillin (100 U/ml), streptomycin (100 Rg/ml) and 10% fetal calf serum (complete medium) at 37Cin the presence of 5% CO 2 2) Preparation of Peptides.
a) Chemical Synthesis.
Peptide synthesis is done by techniques known to a person skilled in the art (Altergen and Neosystem). They are used in solid phase on Fmoc resin. Cleaving is done with trifluoroacetic acid (TFA), and the peptides were purified on a semi-preparatory HPLC-CR C5 column and diluted with a 0.1% TFA solution and an acetonitrile gradient in the TFA. The lyophilized peptides were dissolved in NaCI 0.15 M.
b) Molecular construction allowing preparation of proteins containing the peptides in the invention.
Molecular biology techniques make it possible to construct plasmids which, once introduced into adequate cells, permit the synthesis of vectorized macromolecules.
Construction of vectors for expression of recombinant proteins: Figure 10 attached shows the preparation of vectors that permit the expression of recombinant proteins containing the peptide sequences in the invention. The prokaryotic vector pQE30 (Qiagen) permits the expression of genes in the form of fusion proteins (or recombinant proteins) with the sequence 6XHis.
This vector carries the origin of replication ColEl, the strong promotor of phage T5, which can be induced O by IPTG, the gene for p-lactase giving resistance to ampicillin and a multiple cloning site at 3' of the sequence coding the label 6XHis permitting the cloning of complementary DNA in phase with the 6XHis 00 M sequence.
The complementary oligonucleotides of 63-mer: .PAV1U: in 5'gatccgtaaaacgaggactaaaactacgacacgtagaccacgagtaacacgaatggacgtaa 3' .PAV1L: C\ 5' gatcttacgtccattcgtgttactcgtggtcgtacgtgtcgtagttttagtcctgttttacg-3' are hybridized. The DNA segment obtained has a BamHI site at 5' and a BglII site at It codes (bold letters) for the peptide sequence PAVI: VKRGLKLRHVRPRVTRMDV. This fragment is cloned at the BamHI site of vector pQE30. The complementary DNA (DNAc) coding for the Zebra viral protein (BZLF) of the Epstein-Barr virus (EBV) or the Zebra protein deleted from its nuclear localization site (nls) of 35 amino acids were obtained by PCR. They were cloned at the BamHI site of the vector His- PAV1 or pQE30. The resultant plasmids permit the expression of recombinant proteins Hiss-Zebra-PAVl, Hiss-ZebraAnls-PAVl, His 6 -Zebra and His 6 -ZebraAnls after transformation of the E. coli bacteria.
Induction, extraction and purification of recombinant proteins: The production of recombinant proteins is induced at 37 0 C by the addition of 1 mM of IPTG (isopropyl-p-D-thiogalactopyranoside) to the bacterial cultures in exponential growth phase in Luria Bertani medium supplemented with 40 lg/ml of ampicillin. 12 hours after adding IPTG, the bacteria are centrifuged at 5700 g for 15 min. at 4°C. The bacterial residue is put in 5 volumes of denatured lysis buffer mM Tris-HCI pH 7.8; 0.5 M NaCI; 10% glycerol; 6 M guanidine-HC1). After 20 min. of incubation at ambient temperature with slow mixing, the lysate is clarified by centrifuging it for 30 min. at 15000 g at 4°C. The supernatant containing the recombinant protein is stored at The 6XHis recombinant proteins are purified by affinity chromatography on a "TALON" resin column (CLONTECH) pre-calibrated with denatured lysis buffer. After 3 successive washings of resin with volumes of denatured lysis buffer containing 10 mM imidazole, the recombinant protein bonded to the column is renatured by a gradient of 6 to 0 M of guanidine-HCI in buffer 20 mM Tris-HCI pH7.8; 0.5 M NaCI; 10% glycerol; 0.5 mM PMSF. The recombinant protein is eluted with a gradient of 20 mM at 1 M of imidazole pH 8.0. The different eluates are analyzed on 12% SDS-acrylamide denatured gel. The fractions containing the purified protein are collected and dialyzed for 2 hours at 4 0 C against the buffer 20 mM
I
HEPES pH 7.5, 150 mM NaCI. The protein is concentrated, aliquoted and quick frozen in liquid nitrogen 0 and stored at -80 0
C.
3) Peptides used C a) Non-functionalized.Peptides The following sequences (SEQ ID NO: 1 to SEQ ID NO: 48) are listed in the appendix, in Saccordance with standard SEQ ID NO: 1. Peptide reacting with heparin that comes from the amino acid sequence (3358- 3372) of the human lipoprotein B also called HBP1 below.
SEQ ID NO: 2. Peptide reacting with the heparin dimer of SEQ ID NO: 1, also called (HBP1)2 -below.
0 SEQ ID NO: 3. Peptide reacting with the heparin trimer of SEQ ID NO: 1, also called (HBP1)3 t below.
SEQ ID NO: 4. Peptide corresponding to the hypervariable area CDR3 of the anti-DNA monoclonal murine antibody F4.1 (13).
SEQ ID NO: 5 Peptide containing SEQ ID NO: 1 and SEQ ID NO: 4.
SEQ ID NO: 6. Peptide containing part of CDR2 and CDR3 regions of monoclonal murine antibody F4.1 (13).
SEQ ID NO: 7. Peptide containing SEQ ID NO:1 and SEQ ID NO: 6.
SEQ ID NO: 8. Peptide corresponding to the hypervariable CDR3 region of human anti-DNA monoclonal antibody RTT79 (14).
SEQ ID NO: 9. Peptide containing SEQ ID NO: 1 and SEQ ID NO: 8.
SEQ ID NO: 10. Peptide reacting with heparin and containing SEQ ID NO: 1 and the sequence of the peptide corresponding to the hypervariable area CDR3 of the human anti-DNA monoclonal antibody NE-1 also called No. 1047 below.
SEQ ID NO: 11. Peptide containing SEQ ID NO: 1 and the sequence of the peptide corresponding to the hypervariable area CDR3 of the human anti-DNA monoclonal antibody RT72 (16).
SEQ ID NO: 12. Peptide containing the sequence of the NLS (nuclear localization signal) of cells 3T3 and SEQ ID NO: 6.
SEQ ID NO: 13. Peptide containing SEQ ID NO: 1 and the sequence from CDR2 and CDR3 regions of the anti-DNA human monoclonal antibody NE-1.
SEQ ID NO: 14. Peptide containing part of CDR3 region of murine monoclonal antibody F4.1 and SEQ ID NO: 6.
SEQ ID NO: 15. Peptide containing twice the sequence of the peptide corresponding to the hypervariable CDR3 region of the anti-DNA human monoclonal antibody NE-1.
SEQ ID NO: 16. Peptide resulting from the inclusion, in position 13-19, of SEQ ID NO: 1 in SEQ O ID NO: SEQ ID NO: 17. Peptide reacting with heparin derived from the amino acid sequence of the human lipoprotein E also called HBP4.
I SEQ ID NO: 18. Peptide reacting with heparin derived from the amino acid sequence ofagrine protein of the extracellular matrix that regulates differentiation of the neuromuscular junction.
00 SEQ ID NO: 19. Dimer of SEQ ID NO: 18.
SEQ ID NO: 20. Peptide reacting with heparin derived from the amino acid sequence of "insulin growth factor binding protein" (18).
t SEQ ID NO: 21. Peptide reacting with heparin and derived from the amino acid sequence of the SC-terminal part of chain A of the platelet growth factor also called HPB6.
SEQ ID NO: 22. Peptide containing 12 lysines and SEQ ID NO: 6.
SEQ ID NO: 23. Peptide containing 12 lysines and SEQ ID NO: SEQ ID NO: 24. Peptide having anti-microbial activity (29).
SEQ ID NO: 25. Peptide reacting with heparin and corresponding to the sequence of the insulinlike growth factor-binding protein also called HBP2.
SEQ ID NO: 26. Peptide reacting with heparin and dimer of a peptide derived from the C-terminal part of the sequence of human dismutase superoxide also called (HBP3) 2 SEQ ID NO: 27. Peptide reacting with heparin and corresponding to the sequence SEQ ID NO: 26 in which the amino acids are in configuration D.
SEQ ID NO: 28. Peptide reacting with heparin whose sequence is derived from SEQ ID NO: 26 and contains motif RGD selectively binding the av integrines (21).
SEQ ID NO: 29. Peptide reacting with heparin and composed ofpeptides from SEQ ID NO: 1 and SEQ ID NO: 17, also called HBP1-HBP4 below.
SEQ ID NO: 30. Peptide reacting with heparin and derived from the C-terminal part of the sequence of the epidermal growth factor (EGF) also called HBP7 below.
SEQ ID NO: 31. Peptide reacting with heparin and corresponding to the peptide whose sequence is SEQ ID NO: 12 where the amino acids are in front-back position.
SEQ ID NO: 32. Peptide reacting with heparin and corresponding to sequence SEQ ID NO: 30 in which the amino acids are in configuration D.
SEQ ID NO: 33. Peptide reacting with heparin and containing part of the sequence of the acid fibroblasts growth factor (aFGF) also called HBP8 below.
SSEQ ID NO: 34. Peptide reacting with heparin and containing part of the sequence of the basic Sfibroblast growth factor (bFGF), also called HBP9 (23) below.
(N
SEQ ID NO: 35. Peptide reacting with heparin and corresponding to a C-terminal part of the intestinal murine sequence also called HBP10 below.
SEQ ID NO: 36: Peptide reacting with heparin and containing part of the C-terminal sequence of human y interferon also called HBP11 below.
00 SEQ ID NO: 37: Peptide reacting with heparin and containing part of the sequence of subunit Sof human interleukin 12 also called HBP12 below.
SSEQ ID NO: 38: Peptide reacting with heparin and containing part of the sequence of factor la t derived from stromal cells also called HBP13 below.
1 SEQ ID NO: 39: Peptide reacting with heparin and containing part of the sequence of the "heparin binding protein" (CAP 37) also called HBP15 below.
SEQ ID NO: 40: Peptide reacting with heparin corresponding to the peptide in sequence SEQ ID NO: 10 (1047) plus 13 N-terminal lysines.
SEQ ID NO: 41: Peptide reacting with heparin corresponding to the peptide in sequence SEQ ID NO: 28 (HBP3)2) plus 13 N-terminal lysines.
SEQ ID NO: 42: Peptide reacting with heparin corresponding to the peptide in sequence SEQ ID NO: 39 (HBP10) plus 13 N-terminal lysines.
SEQ ID NO: 43: Peptide with anti-microbial activity containing peptides in sequences SEQ ID NO: 10 (1047) and SEQ ID NO: 24.
SEQ ID NO: 44: Peptide with anti-microbial activity containing peptides in sequences SEQ ID NO: 24 and SEQ ID NO: 30 (HBP7).
SEQ ID NO: 45: Peptide with anti-microbial activity containing peptides in sequences SEQ ID NO: 24 and SEQ ID NO: 38 (HBP13).
SEQ ID NO: 46: Peptide containing the peptide in sequence SEQ ID NO: 26 (HBP3) 2 plus glycine-phthalcyl in N-terminal.
SEQ ID NO: 47: Peptide containing the peptide in sequence SEQ ID NO: 21 (HBP6) plus a salicylyl motif in N-terminal.
SEQ ID NO: 48: Peptide containing the peptide in sequence SEQ ID NO: 21 (HBP6) plus a salicylic motif in C-terminal.
b) Functionalized peptides r 0 These peptides correspond to SEQ ID NO: 1 to 48 above, but carry, on the N-terminal side, either a cystein that allows covalent coupling to some substances of interest (see below) or biotin allowing noncovalent combination of peptides with streptavidin or avidin conjugated with peroxidase (see points and below).
IN II Peptide Penetration Mechanism 1) Involvement of Glvcosaminoglycans (GAG).
0 0 The peptides penetrate the cells by endocytosis mediated by the GAGs present on the membranes Sof all eukaryotic cells. Tests have been done to see their role in peptide penetration.
O a) Heparin (50 ug/ml) was incubated with the cells for 1 hour before adding the peptideperoxidase conjugates at 0.2 ig/ml for 2 hours. The cells were then lysed and the peroxidase dosed.
The 50 jg/ml of heparin totally inhibits the internalization of all peptides in cells H1299 and HeLa.
The quantity of heparin necessary to inhibit 50% penetration of the peptides coupled to peroxidase was evaluated with these two types of cells, by incubating the cells with the peptide-peroxidase conjugates with increasing concentrations of heparin. The results of inhibiting internalization of the peptides are expressed as the heparin concentration (tg/ml) in Table 2 below.
Table 2 Peptide SEQ ID NO: 26 Peptide SEQ ID NO: 21 (HBP6' (HBP3) 2 3T3 cells 14.5 pg/ml 4.1 HeLa cells 8.5 2.3 These experiments demonstrate the role of the heparin sulfates in peptide penetration.
b) The CHO-745 cells are derived from CHO cells. They are deficient in xylosyl transferase, which helps in the formation of the chondroitine sulfate and heparin sulfate of the membrane. The peptideavidin peroxidase complexes do not penetrate into the CHO-745 cells no matter which peptide was tested.
This shows the importance of the heparin sulfate chondroitines in the cell membrane in peptide penetration.
2) Involvement in Cell Metabolism To understand the biological mechanisms whereby the peptides penetrate into the cells and their compartmentalization, experiments are done with drugs whose action on certain cell compartments is specific. The peptide-peroxidases were incubated with H1299 cells at a concentration of 0.2 ig/ml for 2
I
hours in the presence, or not, of a drug. The cells were lysed, and the peroxidase dosed in the cellular lysate.
Table 3 below gives the percentage inhibition of peptide internalization calculated compared to the values given by the lysate of the cells incubated without the drug.
Table 3 Peptide SEQ ID NO: 26 Peptide SEQ ID NO: 30 Peptide SEQ ID NO: (HBP3) 2 (HBP7) Temperature (4 0 C) 100 100 100 Sodium azide 70 70 Sodium chloride 51 69 27 Ammonium chloride 76 70 Genistein (200pM) 59 68 Chloroquin (100pM) 82 79 76 The penetration of the peptides tested is completely inhibited by incubating the cells at 4°C with all peptides. It is partly inhibited by inhibitors of cell metabolism, like sodium azide (inhibitor of ATPase), genistein (inhibitor of tyrosine kinase and of the bond to ATP). The internalization mechanism is therefore dependent on the energy.
The penetration of peptides HBP3 and HBP7 is inhibited on the same order of magnitude by chloroquin, which reflects internalization in the same compartments of the cell cytoplasm (endosomial vesicles, endoplasmic reticulum), while sodium chloride and ammonium chloride (which prevent acidification of the endosomial vesicles, thus inhibiting intracellular traffic) is much less involved in internalization of the peptide in sequence SEQ ID NO: 35. This suggests that the peptides penetrate the cells by mechanisms similar to those used by different toxins, but without having toxicity. The intracellular routes up to the Golgi zone and the retrograde traffic seem distinct, depending on the peptides.
III RESULTS 1) Evaluation of the Capacity of the Peptides to React with DNA and Heparin The capacity of peptides SEQ ID NO: 1 to 21 to attach themselves to DNA and heparin is evaluated on ELISA plates sensitized with DNA or heparin. Dilutions of biotinylated peptides are deposited on the plates and their fixation is shown using streptavidin conjugated with peroxidase. The activity of the peroxidase is revealed with orthodianisidine and H 2 0 2 as a substrate. The avidity of each of the peptides for DNA or heparin is evaluated by the quantity (x 10.6 M) of peptides necessary to obtain 50% fixation. The results obtained are reported in Table 4 below.
Table 4 SEQ ID NO DNA Heparin Chondroitine A Chondroitine B Chondroitine C 1 >100 >100 >100 >100 >100 2 2.2 0.76 0.55 0.6 0.55 3 0.05 0.041 0.037 0.041 0.035 4 4 5 5.6 6.2 0.11 0.11 0.13 0.11 0.11 6 0.19 0.16 0.24 0.25 0.22 7 0.05 0.05 0.06 0.04 0.06 8 >150 >150 >150 >150 >150 9 3.7 3.7 0.017 0.5 >8 3 >30 0.2 4 16 0.019 0.024 0.012 0.009 0.002 17 >150 >150 >150 >150 >150 >134 >74 >.134 >74 >134 >74 >7134 >74 >74 >74 J I I 21 0.06 0.04 I I 4 0.12 0.12 0.12 I_ J_ This table shows that the affinity of SEQ ID NO: 1 for DNA is very small, while that of its dimer (SEQ ID NO: 2) and its trimer (SEQ ID NO: 3) is very great. It also appears that combining SEQ ID NO: 1 with peptic vectors substantially increases the affinity of those vectors for DNA (compare SEQ ID NO: 4 and 5, 6 and 7, 8 and 9, and 15 and 16).
Similarly, the affinity of SEQ ID NO: 17 and SEQ ID NO: 18 for DNA, heparin and the chondroitine sulfates is very small. On the other hand, the affinity for these molecules of the dimer of SEQ ID NO: 18 (SEQ ID NO: 19) is greater. SEQ ID NO: 20 and 21 contain a greater number of amino acids, among which a higher number of basic amino acids have a great affinity for DNA, heparin and the chondroitine sulfates.
The avidity ofpeptides SEQ ID NO: 25 to 39 for various proteoglycanes (Kd x 10- 9 heparin and the chondroitine sulfates is evaluated by the quantity of peptides necessary to obtain 50% fixation on the different antigens. The results are reported in Table 5 below.
Table Peptides Heparin Chondroitine A Chondroitine B Chondroitine C (HBP1) 3 41 37 41 HBP2 70 60 40 100 (HBP3) 2 20 51 43 6
(HBP
3 )2RGD 407 nt nt nt HBP6 16 135 110 12 HBP7 54 78 58 19 HBP8 168 225 260 110 HBP9 45 47 47 HB010 78 112 229 39 HBP11 382 287 301 205 HBP12 295 176 173 93 HBP13 56 56 38 16 100 100 100 100 This table shows that the avidity for heparin of peptides SEQ ID NO: 33 (HBP8), 36 (HBP1 1), and 37 (HBP12) is clearly smaller (>100x10 9 lM) than that of the other peptides, which is on average between 20 and 80x10M. The avidity of the different peptides with chondroitine sulfates A, B and C is, on the whole, on the same order of magnitude as that for heparin. Thus, the avidity of the peptides SEQ ID NO: 34 (HBP9) and SEQ ID NO: 38 (HBP13), for both heparin and the 3 chondroitines is 46-50 x 10" 9
M,
while that of peptides SEQ ID NO: 33 (HBP8), 36 (HBP 11), and 37 (HBP12), for both heparin and the 3 chondroitines, is >100x10 9 M. The modified peptide SEQ ID NO: 28 (HBP3+RGD) lost its affinity for heparin.
2) Evaluation of Penetration of Peptides Inside Cells The cells are cultivated in the presence of the different peptides carrying, on the N-terminal side, biotin in decreasing concentrations (50 to 6 tg/ml) in the culture medium for variable times (1-18 hours).
At the end of the culture, the cells are washed three times with PBS (0.15 M NaCI buffered with 0.01 M potassium phosphate buffer pH 7.4) then fixed 15 minutes in ethanol at -20°C. The penetration of the peptide is evaluated after incubation for 30 minutes with streptavidin conjugated with peroxidase (abbreviated S-PO for streptavidin-peroxidase). Then, the S-PO is eliminated, and the cells are washed. The activity of the peroxidase internalized is revealed by diaminobenzidine in the presence of H 2 0 2 and the cells are examined under a microscope The microscopic examination showed that some of the peptides used (SEQ ID NO: 1, 4,11, 12, 17 and 18) do not give any positive intracellular cytochemical coloration. With the other peptides, positive colorations, but of variable intensity, were noted. Faint coloration was observed with SEQ ID NO: 2, 6, 8 and 19, while, intensive colorations were obtained with the other peptides.
OO
n It is interesting to note that all peptides that contain SEQ ID NO: 1, that is HBP of human lipoprotein B, give a positive coloration except for SEQ ID NO: 1 itself (monomer) and SEQ ID NO: 11.
r N As far as SEQ ID NO: 1, 17 and 18 are concerned, they are short peptides that correspond to the HBP of human lipoproteins B and E and agrine. Unlike those monomer peptides, their dimers (SEQ ID S NO: 2) and especially the trimer of SEQ ID NO: 1 (SEQ ID NO: 3) give clearly positive colorations.
As for SEQ ID NO: 11, it is composed of SEQ ID NO: 1 and CDR3 of anti-DNA human monoclonal antibody RT72 (coded by the germinal line), which contains only one basic amino acid Unlike SEQ ID NO: 11, SEQ ID NO: 5, 7, 9 and 10, which are composed of SEQ ID NO: 1 and CDR2 and/or CDR3 of anti-DNA monoclonal antibodies (coded by the germinal line), contain a high number of basic amino acids and give intense coloration.
On one hand, these results suggest that a minimum of 3 to 5 excess basic amino acids must be present in the peptide sequence so that the peptide is able to penetrate inside the cells. On the other hand, SEQ ID NO: 1 is capable of increasing the translocation power of anti-DNA CDR2 and/or CDR3 only when the latter have a high number of basic amino acids.
The microscopic examination showed that all peptides SEQ ID NO: 25 to 48 give positive cytochemical coloration at a concentration of 6 pg/ml, although the intensity can vary depending on the peptides.
Evaluation of Pentration into Cells of Petide/Streptavidin or Petide/Avidin Complexes Coupled to Peroxidase or Peptide/Peroxidase Conjugates a) A biotinylated/S-PO peptide or biotinylated/A-PO complex (A-PO for avidin-peroxidase) is prepared extemporaneously in two ways: either in a peptide: S-PO or A-PO molar ratio of 4:1 (for example, 1.4 lg of peptide with a molecular weight of 3500 to 10 pg of S-PO; the weight of S-PO used varies depending on the peptides since their molecular weights are different).
or in a peptide: S-PO or A-PO molar ratio of 25:1 (for example 1.7 pg of peptide with a molecular weight of 3500 to 1 Vg of S-PO).
The complexes are prepared by incubating the biotinylated peptide with S-PO or A-PO in PBS in a volume of 5 to 10 tl for 15 minutes at laboratory temperature. They are then diluted in 1 ml of complete VI culture medium, filtered on a 0.2 micron membrane and incubated with the cells for 4 hours. The cells are
O
then washed three times with PBS.
To study the penetration of the complexes under a microscope, the cells are fixed as before for minutes in ethanol at -20 0 C, they are washed with PBS, and the activity of the internalized peroxidase is shown by diaminobenzidine in the presence of H 2 0 2
IO
STo measure the quantity of peptide/S-PO or A-PO complexes internalized, the cells are trypsinized, transferred into microtubes and counted. They are washed twice by centrifugation, then the 00 residue is suspended in 200 pl of lysis buffer (Tris buffer, 0.1 M, pH8, 0.5% Nonidet). After 15 minutes, 0\ the peroxidase contained in the lysis buffer is dosed onto 96-well plates with ortho-dianisidine and H 2 0 2 by comparison with a standard curve for S-PO. The reading is taken at 450 nm. The quantity contained in the Sdifferent samples is expressed in picograms (pg) per 10 4 cells.
Generally, the results obtained by examining the cells under an optic microscope correlate well with the results obtained by measuring the internalized peroxidase. That is the reason that, in what follows, only the quantitative results obtained with a certain number of peptides are given in Table 6 and 7 and Figures 1 to 3.
Table 6 shows an evaluation of the capacity of peptides composed of different HBPs from varied proteins to penetrate H1299 cells, using peptide:S-PO complexes of4:1 and a peptide concentration of 1 Table 6 Table 6 H1299 Cells (pg/10 4 cells) SEQ ID NO: 1 3 SEQ IDNO: 2 84 SEQ ID NO: 3 642 SEQ IDNO: 17 3 SEQ ID NO: 18 3 SEQ IDNO: 19 167 SEQ ID NO: 20 431 SEQ ID NO: 21 1680 The results clearly show that for a vector to be effective in intracellular transfer of the complex S- PO (MW 100,000), a minimum of 6 basic amino acids must be present (SEQ ID NO: 2, 3, 19-21).
Table 7 below reports the results of evaluating the influence of SEQ ID NO: 1 on the penetration of peptides coupled to SEQ ID NO: 1 into H1299 cells, where Concentration a: 25 peptides to one molecule of S-PO (1 pg), Concentration b: 4 peptides to one molecule of S-PO (10 gg).
Table 7 PEPTIDES NOT INCLUDING PEPTIDES INCLUDING SEQ ID NO: 1 SEQ ID NO: 1 4 cells) (pg/10 4 cells) SEQ ID NO: 4 SEQ ID NO: Cone. a 2 Cone. a 100 Conc. b 280 Conc. b 6250 SEQ ID NO: 6 SEQ ID NO: 7 Cone. a 1 Cone. a 240 Conc. b 130 Cone. b 220 SEQ ID NO: 8 SEQ ID NO: 9 Cone. a 50 Cone. a 340 Conc. b 1310 Cone. b 8280 SEQ ID NO: 15 SEQ ID NO: 16 Cone. a 250 Cone. a 430 Cone. b 6670 Cone. b 5220 Table 7 shows that coupling SEQ ID NO: 1 with peptide vectors from anti-DNA human or murine antibodies substantially potentiates the translocation power of those vectors.
Figure 1 is a bar graph illustrating the internalization of streptavidin-peroxidase transported by various biotinylated peptides one night at 37 0 C at concentration a (0.5 nmole of biotinylated peptide to 0.02 nmole of streptavidin-peroxidase).
As can be seen from Figure 1, coupling SEQ ID NO: I with a given vector (SEQ ID NO: 6), which opens onto SEQ ID NO: 7, significantly increases the transfer capacity of said vector. However, coupling a peptide the same length as SEQ ID NO: 1 whose composition of amino acids is close, with this same vector, which opens onto SEQ ID NO: 12, barely increases the translocation power of the vector.
Even coupling a second CDR3 identical to said SEQ ID NO: 6, which opens onto SEQ ID NO: 14, does not result in as sharp an increase as coupling it with SEQ ID NO: 1. Generally, the combination of SEQ ID NO: 1 with vectors of human (SEQ ID NO: 10 and 13) or murine (SEQ ID NO: 5) origin produces particularly effective vectors.
Figure 2 is a bar graph illustrating the internalization of streptavidin-peroxidase (S-PO) and avidin-peroxidase (A-PO) transported by various biotinylated peptides (2 hours at 37 0 C) at concentration b (0.25 nmole of biotinylated peptides to 0.01 nmole of S-PO or A-PO).
SFigure 3 is a bar graph illustrating the ratio between the internalization of S-PO and that of A-PO O by those same biotinylated peptides, 2 hours at 37 0 C, at concentration b (0.25 nmole ofbiotinylated peptide to 0.01 nmole of S-PO or A-PO).
The comparison in Figures 2 and 3 of the transfer of S-PO and A-PO by the vectors suggests that the nature of the substances being transferred influences the translocation capacity of the original allmurine vectors (SEQ ID NO: 14) more than that of the vectors containing HBP of human origin (SEQ ID NO: 7, 10 and 13).
00 00 Figure 4 is a bar graph illustrating the relationship between the internalization of S-PO transported Sby two biotinylated peptides at 37 0 C for 2 hours and at 4°C for 2 hours.
C The results obtained indicate that the translocation power of the vector of human origin (SEQ ID NO: 10) is much more dependent on the temperature (active cell metabolism) than that of the vector of 0 murine origin (SEQ ID NO: 14).
b) To evaluate more directly the penetration of the peptides, avoiding the avidin-peroxidase bias, some of the peptides in sequences SEQ ID NO: 25 to 48 were conjugated covalently with cystein in the C-terminal position of the peptide to peroxidase.
The peptides were coupled with peroxidase in the following way: One mg of peroxidase activated with maleimide (Sigma) in 100 pl of sodium phosphate buffer, NaCI 0.15 M, EDTA 5mM, had 1 mg of peptide added to it. After 2 hours at laboratory temperature, 3mercaptoethanol is added to a final concentration of 1.5 mM for 15 minutes. The uncoupled peptide is eliminated by centrifugation on ultrafiltration membranes (cut-off threshold 30,000 Daltons, Sartorius), followed by three washings in sodium phosphate buffer 0.1 M, pH 7.4 containing 0.15 M NaCI.
The H1299 cells are incubated for 2 hours with the conjugates at increasing concentrations of peptide-peroxidase in the culture medium, then washed with PBS. The cells are lysed and the quantity of peptide internalized is evaluated by the dosage of peroxidase in the lysate of the cells with reference to a curve established with peptide-peroxidase.
The quantity of peptide internalized (pg/10 3 cells), depending on the concentrations of conjugates in the culture medium, is shown in Table 8 below.
Table 8 Concentration (tg/ml) 0.01 0.1 0.5 1 Peptide SEQ ID NO: 28 0.006 1.7 37.9 8860 (0.006)* (8.8) Peptide SEQ ID NO: 34 0.002 1.4 40.7 109 (0.01) (8.14) (10.9) percentage of quantity of peroxidase internalized compared to quantity deposited.
It appears that the higher the concentration of peptide-peroxidase conjugate in the culture medium, the more the quantity internalized increases.
To compare the penetration of the conjugates by cell type, the cells from different lines were incubated with 0.2 ug/ml ofpeptide-peroxidase conjugate for 2 hours, and the quantity ofperoxidase dosed in the lysates. Table 9 and Table 9bis below give the results expressed in pg/10 3 cells. The quantity of each peptide internalized in the different lines generally varies only from single to double.
The internalization ofpeptides HBP3 (SEQ ID NO: 26) and HBP7 (SEQ ID NO: 32) in configuration D is on the same order of magnitude as that of the homologous peptides in configuration L.
The same is true of the peptide in SEQ ID NO: 30 and its homologue in SEQ ID NO: 31, which has an amino acid sequence in front-back position.
Table 9: Internalization of Peptide-peroxidase (pg/103 cells) Cell Lines Peptides 3T3 H1299 HeLa 1047 35 10 4.7 (HBP1) 3 24.6 17.7 11.2 (HBP3) 2 41 27.8 14.9 HBP6 83.7 21.9 13.2 HBP7 48.8 22.5 11 HBP8* 7.6 3.6 2.1 HBP9* 9 3.8 3.4 23.3 7.9 5.3 HBP11 20.3 10.6 4.4 HBP12* 5.7 4.3 HBP13 18.1 9.1 4.1 Table 9 bis: Internalization of peptide-peroxidase 3 cells) Cell Lines Peptides 3T3 H1299 HeLa (HBP3) 2 72.9 67.4 11.4 (HBP3) 2 63.7 59.1 4.1 configuration D HBP7 44.2 63.3 3.6 HBP7 50 56.7 7.7 Configuration D HBP7 front/back 75.9 70.6 8.7 at the limit of the dosage method and consequently significant.
4) Evaluation of penetration ofpeptide-IgG conjugates in cells Two mg of monoclonal IgG antibody or its fragment F(ab')2 in 1 ml of sodium phosphate buffer 0.1M, pH 7 containing 0.15 M NaCI are added to 200 pg of SMCC [succinimidyl 4-(Nmaleimidomethyl)cyclohexane-1-carboxylate] in 10 lI of dimethylsulfoxide, and the solution is incubated for 30 minutes at laboratory temperature. The excess reagent is eliminated by centrifugation on ultrafiltration membranes [cutoff threshold 10,000 Daltons Vivascience], followed by three washings in sodium phosphate buffer 0.1 M, pH 7 containing 0.15 M of NaCl.
The peptide is then coupled in a molar ratio of 6 peptides to 1 IgG in sodium phosphate buffer, 0.1M, pH7, containing 0.15M NaCI for 3 hours at laboratory temperature. Then, the excess uncoupled peptide is eliminated by centrifugation on membranes, as in the preceding step.
To evaluate the penetration of the conjugates under a microscope, the cells are cultivated in the presence of the different antibody-peptide conjugates at decreasing concentrations (50 to 6 tg/ml) in the culture medium for 4 hours at 37 0
C.
At the end of the culture, the cells are washed three times with PBS, then fixed for 15 minutes in ethanol at -20 0 C. The penetration of the IgG-peptide conjugate is evaluated after incubation for 1 hour with an anti-IgG mouse antibody coupled with peroxidase. The cells are then washed with PBS, and the activity of the peroxidase is revealed by diaminobenzidine in the presence of H202.
To measure the quantity of peptide-IgG conjugates internalized, the cells are trypsinized, transferred into microtubes and counted. They are washed twice by centrifugation, then the residue is suspended in 200 pl of lysis buffer (TRIS buffer, 0.1M, pH8, 0.5% Nonidet 40). The quantity of mouse IgG 0 present in the lysate is measured by ELISA on plates coated with mouse anti-IgG sheep antibody and C revealed by a mouse anti-IgG sheep antibody conjugate coupled with peroxidase, with reference to a standard curve established with the same monoclonal antibody that was conjugated with the peptide.
The results are compiled in Table 10, which reports the penetration of murine monoclonal s antibodies or their fragments F(ab')2 inside human H1299 cells, where: a: specific murine monoclonal antibody of protein p53, and 00 b: specific murine monoclonal antibody of protein p21 from a rat-rat hybridoma.
STable Quantity of antibodies Conjugate SEQ ID NO: Conjugate SEQ ID NO: Conjugate SEQ ID NO: Sinternalized 10 IgG 10 F(ab')2 b 10 F(ab') 2 a cells 137 620 383 As can be seen from the table, using vectors of human origin combined with SEQ ID NO: 1 permits effective internalization of the antibody into human cells. Note also that greater quantities of antibodies are transferred into the cells when the vectors are conjugated with F(ab') 2 rather than with the whole antibody.
Figure 5 is a bar graph illustrating the internalization of a murine monoclonal antibody IgGi coupled with different peptides (4 hours at 37C; 30 pg/ml of conjugate deposited) on human H1299 cells and hamster ovary cells CHO.
As can be seen from this graph, the vectors of human origin (SEQ ID NO: 10 and 13) are more effective in transferring substances inside human cells than vectors of murine origin coupled to SEQ ID NO: 1 (SEQ ID NO: 7 and The vectors of human origin (SEQ ID NO: 10 and 13) are also more effective in transferring substances inside human cells than inside hamster cells.
Evaluation of Intracellular Penetration Capacities of Recombinant Proteins by Immunofluorescence.
HeLa cells are sown in a concentration of 0.5 x 10 4 cells on a plate with 24 wells, each containing a sterile lamella. 24 hours later, 50 jg/ml of recombinant protein is deposited on the cells. Six or eighteen hours after that, the cells are washed twice in PBS (phosphate-buffered saline) and fixed with a 4% PFA (paraformaldehyde) solution in PBS for 10 minutes at ambient temperature or dissociated with trypsin and cultured 18 hours on a lamella and then fixed. After three washings in PBS, the cells are permeabilized with a 0.25% solution of TritonX-100 in PBS for 5 minutes at ambient temperature. After three washings in PBS, the cells are incubated for 1 hour at ambient temperature in a moist room with the It monoclonal antibody anti-RGS.His (Qiagen) diluted to 1/50 in PBS 0.1% BSA (bovine serum albumin).
After three 10-minute washings with PBS, the lamella are incubated for 30 minutes at ambient temperature C in a moist room with conjugated goat anti-mouse IgG/FITC diluted to 1/200 in PBS-0.1% BSA. The cells are washed twice in PBS for 10 minutes, then the lamella are mounted on a blade with some mounting liquid (Mounting Medium from Sigma). The cells are observed under a confocal microscope (Leica).
The Hela cells are incubated 18 hours with recombinant protein His 6 -PAV and fixed (Figure 17A) or dissociated and cultured 18 hours before fixation (Figure 17B). Observation of the cells under the 00 confocal microscope shows that the cells incubated 18 hours have major fluorescence on their surface. No
C
r intracellular fluorescence seems to be detected in this experiment. However, contra-coloration of the nuclei by DAPI or HOECHST would make it possible to confirm it. It cannot be ruled out that a small proportion -K of protein is internalized. It also appears that a single PAV1 peptide motif is sufficient to address a recombinant protein on the surface of the cells. After dissociation of the cells, the fluorescence localizes in some endosomal vesicles of different sizes. The recombinant protein His 6 -Zebra-PAV1 is internalized, but does not seem to be nuclear despite the nls domain of the Zebra protein.
The Hela cells are incubated for 6 or 18 hours with the recombinant protein His 6 -ZebraAnls-PAV1 and fixed (Figures 18A and 18B) or dissociated after 18 hours and cultured 18 hours before fixation (Figure 18C). After 6 hours, a nuclear fluorescence is observed that is accentuated at 18 hours. Nuclear areas indicated by arrows have greater fluorescence. For the dissociated cells, more marked fluorescence is located around the nucleoli. Contrary to recombinant protein His 6 -Zebra-PAV1, recombinant protein His 6 ZebraAnls-PAV1 deprived of the nls domain has a nuclear localization. Sequence PAV1 seems to address the protein in the nucleus.
For the Hela control cells (without protein) or the Hela cells incubated with the recombinant proteins Hiss-Zebra and Hise-ZebraAnls, no fluorescence was observed.
6) In vitro evaluation of cvtotoxic activity of ribonuclease A (RNase A) conjugated with pDetides.
mg of bovine RNase A is dissolved in 1 ml of sodium phosphate buffer 0.1M, pH 7 containing 0.15 NaCI.
1 mg of SMCC is dissolved in 50 pl of dimethylsulfoxide (final solution 20 mg/ml).
The RNase A is activated by adding 12.5 1l of SMCC solution into 200 tl of RNase A solution (molar ratio 10 molecules of SMCC to 1 molecule of RNase A).
The reaction takes 30 minutes at laboratory temperature.
The excess reagent is eliminated by centrifugation on ultrafiltration membranes (cutoff threshold 5,000 Da,Vivascience) followed by 3 washings with sodium phosphate buffer 0.1 M containing 0.15 M NaCI.
I
The peptide is then coupled in a 6:1 molar ratio of peptide:RNase A in sodium phosphate buffer O 0.1 M, pH 7, containing 0.15 M NaCI for 3 hours at ambient temperature.
The excess uncoupled peptide is eliminated by centrifugation as in the preceding step.
The quantity of peptide coupled to RNase A is evaluated by the molecular weight of the bands obtained after coupling on an SDS-polyacrylamide gel made of 15% acrylamide.
*The results of electrophoresis comparing RNase A alone (band 1) to RNase A coupled with SEQ ID NO: 10 are shown in Figure 6, where two coupling tests are presented (bands 2 and 3).
o0 Figure 7 shows the results of electrophoresis, used in the same way, to compare the migration of C RNase A alone (band 5) and that of SEQ ID NO: 14 alone (band 6) with that of the product of coupling SRNase A with SEQ ID NO: 14 (two tests: bands 1 and with SEQ ID NO: 5 (band 3) and with SEQ ID 0 (N NO: 10 (band 4).
V' RNase A coupled with vectors of human origin (SEQ ID NO: 10, 15 and 16) was tested for its O cytotoxic activity on HH9 cells in culture by comparison with natural RNase A.
a) In vitro Evaluation of Cvtotoxic Activity of RNase on HH9 Cells.
The cells are sown at night on plates with 96 wells, at the rate of 10 3 cells per well.
The next day, the supematant is suctioned off and replaced with 100 gl of medium containing succession dilutions of RNase A-vector or natural RNase A and the culture continued for 72 hours.
The wells are then filled with 50 tl of a solution of MTT (3-(4,5-dimethylthiazol-2-yl)-2,5diphenyl tetrazolium bromide) at 1 mg/ml in the culture medium and cultivated for 4 hours.
The supernatant is removed, and 100 Vl of dimethylsulfoxide is added to the wells. After the crystals dissolve, the coloration is read at 550 nm.
The results are calculated as a percentage of the average optic density obtained in the wells of cells containing the dilutions of the samples being tested and the optical density of the wells that received only medium. These results are shown graphically in Figure 8.
As can be seen from Figure 8, natural RNase A has no cytotoxic activity. The average cytotoxicity of 50% is obtained at concentrations of RNase A-vector between 33 and 100 pg/ml, between the molar concentrations of 2 to 7x10"-M.
b) In vitro Evaluation of Cvtotoxic Activity of Peptide-RNase Conjugates on Hela Cells The different peptide-RNase conjugates were tested on varied cell lines. The results are expressed as IC50, which corresponds to the concentration of the peptide-RNase conjugate giving 50% inhibition of cell growth after 72 hours of culture.
Table 11 summarizes the results obtained. It appears that all the conjugates have distinct cytotoxic activities and that some of them have little cytotoxic power.
O
O
IN
0 0 0 OIn o- Table 11 Cell Lines Peptide- HT29 H1299 HH9 HUVEC 3T3 MCF7 HeLa B16.F10 RNase (HBP1)3- 70 100 20 80 40 90 60 (HBP1)2- nt* 25 nt nt nt nt nt nt HBP1-4- nt 25 nt nt nt nt 6 nt HBP 3- 10 12 2 15 3 15 7 12 HBP6- 45 3 3 50 3 20 12 nt HBP -7 45 50 2 50 6 25 15 nt HBP 8- nt nt nt nt nt nt >100 nt HBP9- nt nt 100 nt nt nt >100 nt HBP 10- >100 150 33 >100 50 150 25 nt HBP 11- >100 >100 60 >100 60 >100 20 nt HBP 12- >100 >100 >100 >100 40 >100 >100 nt HBP 13- 40 100 8 65 14 >100 3 nt HBP 14- >100 250 >100 >100 55 90 25 nt HBP >100 250 60 >100 55 90 30 >100 1047- *nt: no 7) In vivo Evaluation of Cvtotoxic Activity of RNase A Conjugated with Peptides on Human Tumors Grafted onto Athvmic (Nude) Mice a) Protocol 1.
Female nude mice 6 weeks old were injected subcutaneously with 3x 06 HH9 cells, volume 50 Al, in the left side. On day 17 after the transplant, the tumors were measured, and the mice were divided into 3 groups: Group 1 (7 mice) injected with 100 lig of a coupling SEQ ID NO: 10/RNase A in 50 1 l of PBS, Group 2 (7 mice) injected with 50 jg of natural RNase A in 100 pl of PBS, Group 3 (6 mice) injected with pl of PBS. The injections were given three times a week under the same conditions. Each time, the tumors were measured, and their volume was calculated using the formula V 4/37xL21 where L is for the large diameter of the tumor and 1 for the small diameter of the tumor.
The results are shown graphically in Figure 9.
As the figure shows, after the 3 3 rd day, the progression of the volume of the tumors is clearly slowed by the RNase A coupled with SEQ ID NO: It would seem that there is less cytotoxicity for some tumor cells than for others.
b) Protocol 2.
The nude mice received subcutaneous transplants of HH9 tumor cells as described above. On day 9 after the transplant, the mice received twice a week, by peritumoral injection, 100 pg ofRNase, or 65 pg of peptide (HBP3) 2 or 65 pg of peptide (HBP3) 2 100 pg of RNase, or 100 pg of peptide conjugate (HBP3) 2 -RNase or HBP6-RNase conjugate. On day 26, the mice were killed and their tumors weighed.
The inhibition of the growth of the HH9 tumors treated with the peptide-RNase conjugates is given in Table 12. The inhibition of the growth of the tumors of the mice treated is calculated in relation to the average weight of the tumors of mice that received NaCI.
Table 12 RNase NaCI (HBP3) 2 (HBP3) 2 (HBP3) 2 HBP6- HBP7group +RNase RNase RNase RNase Tumors* 1720 1560 1500 790 810 830 Inhibition** 10 13 54 53 52 Average weight of tumors of 6 mice in the same group in mg.
The 3 peptide-RNase conjugates inhibit the growth of tumors in an equivalent way, while the peptide alone or with RNase added has little effect.
8) In vitro Transfection of Cells with Plasmid of Luciferase Using Peptides a) Transfection of CHO Cells.
The plasmid used is PCMV-LUC (6.4 kb) carrying the luciferase gene under the control of the promotor of the human cytomegalovirus.
The plasmid-peptide complexes are prepared by adding 3 pg of plasmid to 50 pl of NaCl, 0.15 M and 3.3 nmoles of SEQ ID NO: 22 and 23 for 15 minutes. The complexes are prepared in triplicate.
CHO cells are cultivated in the complete medium MEM Alpha containing 6% fetal calf serum and sown with 5x10 4 cells per well on a 24-well plate the evening before the experiment. The wells are then emptied of the medium, and the plasmid-peptide complex is added to them (3 pg of plasmid and 3.3 nmoles of peptide in 0.5 ml of complete medium for 5 hours at 37 0 The medium is then replaced with the complete medium, and the culture is continued for 18-20 hours. The cells are then washed three times with PBS, then lysed with 100 pl of lysis buffer (Promega) containing 1% Triton X-100 and 2 mM DTT.
Then a 15 pl sample of the lysate is taken to measure the luciferase with the Promega luciferine kit and 15 other pl to calculate the quantity of proteins in the lysate using the Bradford Test. The relative luciferase units (RLU) are reported in mg of proteins.
The results are reported in Table 13 below.
Table 13 RLU 10 6 /mg of proteins SEQ ID NO: 22 1.9 IN SEQ ID NO: 23 11.5 The results show that the expression of luciferase is possible by complexing the plasmid with one 00 f< of the vectors and that the expression is increased 6 times (SEQ ID NO: 23) by adding HBP of apolipoprotein to SEQ ID NO: 22.
Sb) Transfection of 3T3 Cells I) Technique: The plasmid pCMVLUC (carrying the gene for luciferase) provided by Qiagen is used according to their instructions. 3T3 cells are sown the night before in 24-well plates (8x10 4 cells/well) in the complete culture medium.
The peptides and the plasmid are complexed in a ratio of 1.6 nmoles/pg in 50 ul ofNaCl 0.15M for 20 minutes at laboratory temperature. The cells are then washed, and the peptide-plasmid complex added to the cells in 0.5 ml of complete medium. After 5 hours of incubation at 37 0 C, the reaction medium is eliminated, and 1 ml of complete medium is added to the cells. After 24 hours of culture, the cells are then washed 3 times with PBS and lysed with 100 pl of lysis buffer (Promega) containing 1% Triton X-100 and 2mM DTT for 15 minutes. The luciferase test is done on the cell lysate after centrifugation. The quantity of protein in the lysate is measured using the Bradford Test (Bio-Rad) and calculated according to a curve established with bovine gamma globulins. The luciferase activity is estimated on 15 pl of lysate plus 100 pl of reagent of luciferase containing luciferine (Promega). The relative luciferase units (RLU) are measured in a luminometer (Berthold Systems) and reported in mg of protein.
Results: The quantity of luciferase expressed by the transfected cells is shown in Table 14 below.
Table 14 pCMVLUC alone Peptide SEQ ID Peptide SEQ ID Peptide SEQ ID NO:41 NO:42 RLU 0.004* 1.9 0.9 x 10 6 /mg of protein The peptide complexed to the plasmid permits its transfer inside the cells as well as expression of the gene for luciferase. The efficacy of the transfection expressed by the quantity of luciferase (RLU/mg of protein) varies depending on the peptides used. Peptide SEQ ID NO: 42 and peptide SEQ ID NO: 40 give values 5.5 times and 2.6 times higher, respectively, than peptide SEQ ID NO: 41.
9) Evaluating the Penetration of Substances using Transporters Coupled to Peptides If we now take the transporters in the invention, Table 15 below gives some examples of transporters and substances of interest for which they have a strong affinity.
Table TRANSPORTERS SUBSTANCES OF INTEREST Peptide Protein A Human or rabbit IgG Peptide Protein G Bovine, goat or mouse IgG Peptide anti-IgG F(abl)2 All IgG from the same species Peptide anti-peroxidase IgG Peroxidase and any molecule containing peroxidase Peptide anti-RNase F(abl)2 Ribonuclease A Peptide Concanavalin A A large number of glycoproteins Peptide Streptavidin-Avidin Biotin and any molecule containing biotin In the table above, the general term "peptide" means "sequence of amino acids in the invention." Protein A is a protein isolated from Staphylococcus aureus which has a molecular weight of 42,000 Da. The characteristic property of this protein is its great affinity for the Fc part of the IgG. This property makes it possible for it to attach 2 to 4 molecules of IgG without interacting with the active sites of the antibodies. Protein A is interesting for human, rabbit and rat IgG, and some mouse IgG.
Protein G, originally, was a protein from 30,000 to 35,000 Da isolated from thecell wall of bacterial strains C or D of streptococcus. Protein G sold by SIGMA is a recombinant protein of 17,000 Da that contains two IgG fixation domains. Like protein A, protein G has a strong affinity for the Fc fraction of the IgG, but it will be used mainly for bovine, mouse and sheep IgG.
F(ab')2 come from antibodies. We will use as examples mouse anti-IgG sheep IgG obtained from sheep immune serum. They are purified by affinity chromatography in which the solid phase is composed of mouse IgG fixed on spheres of polyacrylamide-agarose. This is purification by immunoabsorbent that permits specific fixation of the mouse anti-IgG sheep IgG by an antigen-antibody reaction. This antigenantibody bond is dissociated by an acid pH. Managed digestion of the IgG by proteolytic enzymes makes it possible to obtain fragments of different antibodies. Pepsin is used to obtain bivalent fragments F(ab') 2 reduced in size (MW=92,000 Da), and Fc fragments totally digested into small polypeptides. Eliminating the Fc fragment makes it possible to reduce the size of the antibodies and simplifies coupling with the peptides. Creating an anti-IgG peptide-F(ab')2 transporter will make it possible to internalize a great variety of mouse IgG in large quantities.
Anti-peroxidase IgG is a monoclonal antibody of isotype IgGI that is isolated from mouse ascites.
It is purified by affinity chromatography on protein G (GammaBind Plus Sepharose: Pharmacia). This antibody has a strong affinity for peroxidase and, by the same token, for all molecules containing peroxidase. Peroxidase (PO) is an enzyme extracted from the black radish with a molecular weight of 44,000 Da, which can be combined very easily with many molecules. Creating an anti-PO peptide-IgG will make it possible to internalize PO and any substance containing PO into the cells.
Concanavalin, extracted from Canavalia ensiformis, is a lectin that has an affinity for the terminal 00 groups a-D-mannosyl and ac-D-glucosyl of proteins. This property allows it to react with many glycoproteins. Creating a peptide-concanavalin transporter will make it possible to internalize many glycoproteins or glycolized molecules into cells.
Streptavidin, a molecule weighing 60,000 Da extracted from Streptomyces avidinii, and avidin, Sextracted from egg white, both have four biotin recognition sites. Their affinity constant for this small molecule is very high (KD 10" 1 M) which allows them to interact with all substances containing biotin.
Creating a peptide-streptavidin or peptide-avidin transporter will make it possible to internalize biotin and any molecule containing biotin into the cells.
The examples described below illustrate the creation and use of such transporters.
a) Evaluating the Penetration of Bovine Ribonuclease A (RNase A) Transported by a Transporter peptide-F(ab')?. anti-RNase into the Cells Managed digestion of the anti-RNase IgG by a proteolytic enzyme, pepsin, makes it possible to obtain fragments F(ab') 2 This digestion, which is done in a sodium acetate solution pH 4.5 with 2% pepsin, makes it possible to fragment the fragment Fc of the IgG without altering the active sites of the antibody.
Two mg of antibody F(ab') 2 anti-RNase in 1 ml of potassium phosphate buffer 0.1 M pH 7.4 are added to 110 lg of SMCC [succinimidyl 4-(N-maleimidomethyl)cyclohexane-l-carboxylate] in 11 l of dimethylsulfoxide. The solution is incubated for 45 minutes at laboratory temperature. The excess reagent is eliminated by centrifugation on ultrafiltration membranes (cutoff threshold 10,000 Da, Vivascience), followed by three washings in sodium phosphate buffer 10 mM, pH 7 containing 01.5 M of NaCI and 5 mM of EDTA.
The coupling with the peptide is then done in a molar ratio of 6 peptides to 1 F(ab') 2 in the sodium phosphate buffer 10 mM, pH 7, containing 0.5 M of NaCI and 5 mM of EDTA for three hours at laboratory temperature. Then, the excess uncoupled peptide is totally eliminated by centrifugation on ultrafiltration membranes, as in the preceding step.
These transporters [anti-RNase peptide-F(ab') 2 are added to ribonuclease A in some complete culture medium, and the solution is incubated for 1 hour at laboratory temperature. The molar ratio of this reaction is 2 RNase/1 F(ab') 2 To evaluate the penetration of RNase A under the microscope, the cells are cultivated in the presence of the complexes [anti-RNase peptide-F(ab') 2 ]-RNase A diluted to decreasing concentrations of RNase A (from 100 to 6 pg/ml) in some culture medium for 4 hours at 37 0
C.
At the end of the incubation, the cells are washed three times with PBS, then fixed for 15 minutes in absolute ethanol at -20 0 C. The penetration of the RNase is evaluated after incubation for 1 hour with an anti-RNase antibody coupled with peroxidase. The cells are then washed with PBS, and the peroxidase activity is revealed by diaminobenzidine in the presence of H 2 0 2 To measure the quantity of RNase A internalized, the cells are trypsinized, transferred into microtubes and counted. They are washed twice by centrifugation with PBS, then the residue is suspended in 220 til of lysis buffer (tris buffer, 0.1 M, pH 8, 0.5% Nonidet 40). The quantity of RNase A present in the cell lysate is measured by ELISA on plates sensitized with anti-RNase A rabbit antibody and revealed by an anti-RNase A rabbit antibody conjugate coupled with peroxidase, with reference to a standard curve established with RNase.
The results given in Table 16 illustrate the internalization of RNase A by the anti-RNase transporter F(ab') 2 and the internalization of the RNase coupled covalently to the peptide inside the HeLa cells.
Table 16 Transporter Peptide- Natural RNase A RNase A RNase Quantity of RNase internalized (pg/10 4 476 433 0 cells) b) Evaluation of Penetration of Mouse IgG Transported by a peptide-F(ab') mouse anti-IgG Transporter Inside Cells The peptide-F(ab') 2 mouse anti-IgG transporters are obtained by the technique described above.
These transporters [peptide-F(ab')2 anti-IgG] are added to the mouse IgG in some NaCI buffer. The solution is incubated for 1 hour at laboratory temperature. The molar ratio of this reaction is 1 F(ab')2/1 IgG. After an hour of incubation, the complexes formed are diluted in some complete culture medium.
To evaluate the penetration of the IgG under the microscope, the cells are cultivated in the presence of complexes [peptide-F(ab')2 anti-IgG]-IgG diluted to decreasing concentrations of IgG (from 100 to 6 ug/ml) in some complete culture medium for 4 hours at 37 0
C.
At the end of incubation, the cells are washed three times with PBS, then fixed for 15 minutes in absolute ethanol at -20 0 C. The penetration of the IgG is evaluated after incubation for 1 hour with an anti- IgG antibody coupled to peroxidase. The cells are then washed with PBS, and the activity of the peroxidase is revealed by diaminobenzidine in the presence of H 2 0 2 tI To measure the quantity of IgG internalized, the cells are trypsinized, transferred into microtubes 0and counted. They are washed twice by centrifugation with PBS, then the residue is suspended in 220 pl of lysis buffer (tris buffer 0.1M, pH 8, 0.5% Nonidet 40). The quantity of IgG present in the cell lysate is Smeasured by ELISA on plates sensitized with anti-IgG sheep antibody and revealed by an anti-IgG sheep antibody conjugate coupled with peroxidase, with reference to a standard curve established with the IgG.
O The activity of the peroxidase is revealed with orthodianisidine and H 2 0 2 The same transporter [peptide-F(ab') 2 anti IgG] can transport a large variety of mouse IgG inside the cells.
C The results given in Table 17 illustrate the internalization of different mouse IgG or potentially rat IgG inside HeLa cells via the same transporter [peptide-F(ab') 2 anti-IgG].
t Table 17 Quantity of IgG internalized 4 cells) Transporter anti-p53 IgG 1100 Transporter anti-p21 IgG 1200 Transporter anti-PO IgG 695 Transporter biotinylated IgG 153 Transporter mouse IgG 5960 Transporter F.4.1 3200 F.4.1 1299 Anti-p53 IgG: specific murine monoclonal antibody of protein p53.
Anti-p21 IgG: specific monoclonal antibody of protein p21 that comes from a rat-rat hybridoma, reacting with mouse anti-IgG antibodies.
Anti-PO IgG: specific murine monoclonal antibody of peroxidase.
F.4.1: anti-DNA murine monoclonal antibody that penetrates inside cells by itself.
c) Evaluation of Penetration of Peroxidase (PO) or Molecules Containing Peroxidase Transported by a peptide-IgG anti-PO Transporter into Cells Two mg of specific monoclonal IgG antibody of peroxidase or their fragment F(ab') 2 in 1 mi of potassium phosphate buffer 0.1 M, pH 7.4 are added to 110 lg of SMCC [succinimidyl 4-(Nmaleimidomethyl)cyclohexane-1-carboxylate] in 11 pl of dimethylsulfoxide. The solution is incubated for minutes at laboratory temperature. The excess reagent is eliminated by centrifugation on ultrafiltration membranes (cutoff threshold 10,000 Da, Vivascience), followed by three washings in sodium phosphate buffer 10mM, pH 7 containing 0.5 M of NaCl and 5 mM of EDTA.
The peptide is then coupled in a molar ratio of 6 peptides to 1 IgG or 1 F(ab') 2 in the sodium phosphate buffer 10 mM, pH 7, containing 0.5 M of NaCI and 5 mM of EDTA for three hours at laboratory temperature. Then, the excess uncoupled peptide is totally eliminated by centrifugation on ultrafiltration membranes, as in the preceding step.
These transporters [peptide-IgG anti-PO] are added to the peroxidase or the biotinylated peroxidase in some 0.5 M NaCI buffer. The solution is incubated for 1 hour at laboratory temperature. The molar ratio of this reaction is 2 PO 1 IgG or F(ab') 2 00 To evaluate the penetration of the PO or the molecules containing PO under the microscope, the c cells are cultivated in the presence of [peptide-IgG anti-PO]-PO complexes at decreasing concentrations of PO or molecules containing PO (100 to 6 gg/ml) in some culture medium for 4 hours at 37°C.
O
fC~ At the end of the culture, the cells are washed three times with PBS, then fixed for 15 minutes in t absolute ethanol at -20 0 C. The cells are then washed with some PBS, and the penetration of the PO or the molecules containing PO is evaluated by the activity of peroxidase revealed by diaminobenzidine in the presence of H202.
To measure the quantity of PO internalized, the cells are trypsinized, transferred into microtubes and counted. They are washed twice by centrifugation with PBS, then the residue is suspended in 220 pl of lysis buffer (tris 0.1 M buffer, pH 8, 0.5% Nonidet 40). The quantity of PO present in the cell lysate is measured by ELISA on plates sensitized with anti-peroxidase mouse antibody and revealed with orthodianisidine and H202 with reference to a standard curve for PO.
The results given in Table 18 illustrate the internalization of the PO and the biotinylated PO inside HeLa cells via the transporter [peptide-IgG anti-PO].
Table 18 Peroxidase (PO) Biotinylated PO Quantity of molecules internalized (pg/10 4 cells) 2100 1700 d) Evaluation of Penetration of IgG Transported by a Peptide-protein A Transporter into Cells One mg of protein A in 0.5 ml of sodium phosphate buffer 0.1 M, pH 7, is added to 120 pg of SMCC [succinimidyl 4-(N-maleimidomethyl)cyclohexane-l-carboxylate] in 12 pl of dimethylsulfoxide.
The solution is incubated for 45 minutes at laboratory temperature. The excess reagent is eliminated by centrifugation on ultrafiltration membranes [cutoff threshold 10,000 Da, Vivascience], followed by three washings in sodium phosphate buffer 0.1 M, pH 7 containing 0.15M of NaC1.
The peptide is then coupled in a molar ratio of 6 peptides to 1 protein A in the sodium phosphate buffer 0.1 M, pH 7, containing 0.15 M of NaCI for three hours at laboratory temperature. Then, the excess uncoupled peptide is totally eliminated by centrifugation on ultrafiltration membranes, as in the preceding step.
V These transporters [peptide-protein A] are added to some IgG in some 0.15 M NaCI buffer. The Ssolution is incubated for 20 minutes at laboratory temperature. The molar ratio of this reaction is 2 IgG/1 protein A.
To evaluate the penetration of the IgG under a microscope, the cells are cultivated in the presence of [peptide-protein A]-IgG complexes diluted at decreasing concentrations of IgG (100 to 6 pg/ml) in some IN culture medium, for 4 hours at 37 0
C.
At the end of the culture, the cells are washed three times with some PBS, then fixed for minutes in absolute ethanol at -20 0 C. The penetration of the IgG is evaluated after incubation for 1 hour 00 with an anti-IgG antibody coupled with the PO or with PO alone for the anti-PO IgG. This step is not necessary in the internalization of IgG conjugated with PO (for example: rabbit IgG-PO). The cells are then Swashed with PBS, and the activity of the peroxidase is revealed by diaminobenzidine in the presence of in H 2 0 2 STo measure the quantity of IgG internalized, the cells are trypsinized, transferred into microtubes and counted. They are washed twice by centrifugation with PBS, and then the residue is suspended in 200 pl of lysis buffer (tris buffer 0.1M, pH 8, 0.5% Nonidet 40). The quantity of IgG present in the cell lysate is measured by ELISA on plates sensitized with anti-IgG antibody and revealed by an anti-IgG antibody conjugate coupled with peroxidase, with reference to a standard curve established with the IgG. The PO activity is revealed with ortho-dianisidine and H 2 0 2 The results given in Table 19 illustrate the internalization of different IgGs inside HeLa cells via the transporter [peptide-protein A].
Table 19 Rabbit IgG-PO Mouse IgG IVIg Anti-PO Quantity of IgG internalized (pg/10 4 10800 480 26000 cells) Transport of anti-CEA Antibody in Colon Cancer Cells a) Experiments were done with anti-CEA 35A7 Ab and the peptides in sequence SEQ ID NO: SEQ ID NO: 30 and SEQ ID NO: 35, also called 1047, HBP7 and HBP10, respectively.
Peptide 1047 was coupled with 35A7 and Herceptin®. After a test of the antibody activities of the conjugates by ELISA, the internalization was studied by immunofluorescence in comparison with the initial antibody at 37 0 C and 4°C. On cells LS174T, human colon carcinoma CEA+, the conjugate 35A7- 1047 internalizes after 1 hour 15 minutes, while 35A7 showed no internalization until 5 hours. On SKOv3 cells, human ovarian carcinoma ErbB2+, the conjugate Herceptin®-1047 presents rapid internalization close to that obtained with Herceptin®. After radiomarking with 125I, the conjugates demonstrate 0 immunoreactivity close to the initial Ab on Ag immobilized on Sepharose. A gel filtration analysis shows the absence of aggregate.
b) Comparison of peptides 1047, HBP7 and HBP10. This comparison concerns only Ab 35A7.
S- In vitro experiments A kinetic study of the internalization of different conjugates (35A7-1047, 35A7-HBP7 and 35A7- HBP10) on LS174T cells shows greater internalization with peptide HBP7 than with 1047 and These two peptides give comparable results. On SKOv3 and CEA- cells, 1047 and HBP7 induce OO internalization comparable to that obtained on LS174T, while HBP10 gives results cut in half.
In vivo experiments The 3 conjugates 35A7-1047, 35A7-HBP7 and 35A7-HBP10 marked with 125 I were studied on K1 nude mice with LS174T tumors compared with marked 35A7. 1 1-35A7 shows around 13% tumor capture of the dose injected per gram 6 hours post-injection. This capture is between 20% and 25% ID/g of 0tumor 24 hours post-injection. Conjugate 1 25 I-35A7-1047 demonstrates somewhat faster elimination and slightly less tumor capture compared to '"I-35A7 (18.6% vs. 21.2% ID/g at 24 Conjugate 1 25 I-35A7- HBP7 shows major hepatic capture after 6 h. This causes rapid elimination and low tumor capture ID/g at 24 Conjugate 125I-35A7-HBP10, with a biodistribution very close to that of the natural antibody, has the best tumor capture of the 3 conjugates.
c) The induction effect of internalization with peptide 1047 is particularly visible on 35A7, an Ab directed against CEA which is an Ag that does not internalize. The effect is less evident on Herceptin®, which internalizes quickly after fixation to its Ag, ErbB2.
The in vitro experiments done with 1047, HBP7 and HBP10 show that the three peptides induce internalization of 35A7. HBP7 seems to be the most active, but HBP10 is the one that seems to respect most the specificity of the antibody (marked effect on CEA+ cells, reduced effect on CEA- cells). The in vivo experiments confirm the results obtained in vitro.
d) Figures 11 to 16 attached illustrate the results above.
The peptides: S1047: 2420 Da on anti-ACE and anti-erbB2 Ab HBP7: 1827 Da on anti-ACE Ab only HBP10: 1696 Da on anti-ACE Ab only Coupling: After refinement, the conditions set for coupling (preventing the formation of antibody aggregates) are: 18 SMCC/Ig and 12 pept/Ig-SMCC Internalization test on cells in cultures: SFigures 11A and B show Test 1: on A375 cells (negative ACE) and 5F12 cells (positive ACE), 40,000 cells per well (24-well box). Incubation with natural antibody 35A7 versus conjugated Ab: 35A7-1047, 4 hours at 37 0 C. Washing, fixation, incubation with secondary-peroxidase Ab, OPD indicator, reading 490 nm. DO based on concentration of antibody in Ig/ml.
Vt Figure 12 shows Test 1 with the antibodies marked with iodine 125.
O
Figures 13A and B show Test 2: on A375 cells (negative erbB2) and SKOV3 cells (positive erbB2), 40,000 cells per well (24-well box). Incubation with natural antibody Herceptin versus h conjugated Ab: HER-1047, 4 h at 37 0 C. Washing, fixation, incubation with secondary-peroxidase Ab, OPD indicator, reading 490 nm.
IN Figures 14A and B show Test 3: by reducing the concentration of antibodies and varying the incubation time 1 h or 4 h.
Figures 15A to F show Test 4: identical protocol but by comparing the three peptides (1047- 00 H HBP7-HBP10) on SKOV3 (ACE-) and LS174T (ACE+) cells.
Figures 16A to F illustrate the in vivo experimentation: distribution of antibodies 35A7-1047, S35A7-HBP7, 35A7-HBP10 radiomarked with iodine 125, versus 35A7-iodine 131; Swiss nude/nude mice t) implanted with LS174T (human colon carcinoma, CEA+); co-injection of 5 pg Ab-pept* and 5 pg of Snatural Ab* per mouse, by dissection 6h 24h post-injection, expression of the results as a percentage of the dose injected per gram of tissue. Black bar: conjugated Ab 1251 white bar: natural Ab 1311.
11) Transport of Particles Using a Peptide-IgG Derivative The vector used in the following examples is a monoclonal mouse antibody (IgG1) coupled with the peptide in sequence SEQ ID NO: 10 (1047). The peptide was coupled with the monoclonal antibody as described above.
a) Fluorescent Spheres: Preparation of peptide IgG microsphere complex: Some 70-900nm fluorescent polystyrene microspheres carrying anti-IgG mouse antibodies on their surface (Estapor, Merck eurolab) are diluted to 1/100 in NaCI 0.15M. 4 pl of this preparation is added to a volume of 50 pl containing 10 pg of monoclonal mouse IgG conjugated with peptide SEQ ID NO: 2 and left to incubate for 30 minutes at laboratory temperature. This reaction medium is deposited on the HI299 cells sown (5x10 4 cells/well) the night before for 18 hours.
The cells are washed and observed under a fluorescence microscope: the cells incubated with the fluorescent peptide-IgG sphere complexes are highly marked, whatever the size of the spheres, while the controls (fluorescent spheres without peptide-IgG, or with normal IgG) are negative.
Colloidal Gold: preparation of peptide-IgG-colloidal gold complex: Spheres of colloidal gold (British Biocell International) of 10 nm are complexed to monoclonal mouse IgG, conjugated beforehand with peptide 1047, according to the manufacturer's instructions in a ratio of 750 pg of IgG per 1 ml of colloidal gold solution. For the penetration of the complex into the cells, the residue of the suspension is diluted in half in the culture medium.
c) Evaluation of Internalization of Peptide-IgG Spheres Complexes fluorescent and colloidal gold under the electron microscope.
After the conjugation steps, the preparations are deposited on H1299 cells cultivated since the night before as described above. After 18 hours of culture for the IgG-fluorescent spheres complex or 4 hours for the IgG-colloidal gold complex, the cells are washed with PBS three times, then fixed in a 1.6% 0 glutaraldehyde solution in sodium phosphate buffer, 0.1M for one hour, then treated as usual for the 0 electron microscope.
Results: The peptide IgG-microsphere complexes of 70 nm (Figure 19) and the peptide-IgGcolloidal gold complexes (Figure 20) are visible in some vesicles and in the endoplasmic reticulum of the cytoplasm and around the Golgi area.
12) Transport of Doxorubicin.
Anthracyclines like doxorubicin are among the most active agents in the treatment of human 00 cancers. Their mode of entry into the cells is not yet resolved. What is known is that they are quickly Stransported into the nucleus of the cells. Their toxicity, probably due to their broad diffusion through the body, is a limitation in treatment, preventing the use of a large quantity.
NC1 We wanted to see if coupling doxorubicin to the HBP peptides would improve its power to inhibit the growth of human tumors while reducing the toxicity of the drug.
Sa) Coupling Peptides to Doxorubicin Doxorubicin, whose amino group is protected, is treated with 4-maleimidobutyric acid in the presence of carbodiimide, which results in the formation of an ester bond between the hydroxyl of doxorubicin and the carboxyl of maleimidobutyric acid. The maleimide group introduced into doxorubicin reacts with the cystein present in terminal -C or -N of the peptide.
The following conjugates were prepared following the general plan:: 1047 doxorubicin; HBP 1-doxorubicin; HBP3-doxorubicin; HBP6-doxorubicin; HBP7doxorubidin; HBP10-doxorubicin; HBP13-doxorubicin.
b) In vitro Evaluation of the Biological Activity of Doxorubicin Coniugated with Peptides.
To evaluate the biological activity of peptide-doxorubicin conjugates, the inhibition of the growth of tumor cells in culture is measured.
The cells are sown the night before in 96-well plates at the rate of 10 3 cells per well. The next day, the supernatant is suctioned off and replaced by 100 pl of medium containing successive dilutions of peptide-doxorubicin or natural doxorubicin (10- 6 M 10 8 M) and the culture continued for 48 hours.
Then, the wells are filled with 50 pl ofMTT (3-(4,5-dimethylthiazol-2yl)-2,5-diphenyl tetrazolium bromide) at 1 mg/ml in the culture medium and cultivated for 4 hours. The supernatant is removed, and 100 pl ofdimethylsulfoxide is added to them. After the crystals dissolve, the coloration is read at 550 nm.
The results are calculated as a percentage of the mean optical density obtained in the wells of cells containing the dilutions of the samples being tested and the optical density of the wells that received only medium. They are expressed in a molar concentration giving 50% inhibition of the growth of the cells and are reported in Table 20 below.
Table Cell Lines H1299 HH9 HeLa B16.F10 K562 K562R* MCF7 MCF71 Doxorubicin 3x10' -3x10' 2x10 x10 4xl0 3xl0 2x10 2x10
\O
(natural)
(HBPI)
3 7x10' nt* nt* nt* 2x10 5x0 6x10' IxI 00 doxo M (HBP3) 2 nt* nt* nt* nt* x10 I- 3x10 I 5x10 7 2.5x10 doxo HBP6-doxo nt* nt* nt* nt* 3x10 5x10 6x10 6.5x10 HBP7-doxo nt* nt* nt* nt* Ix10 1x10 7 7x10 4x10 HBPl0-doxo nt* nt* nt* nt* 8x10- 1x10 5x10 3.5x10 HBPI3-doxo nt* nt* nt* nt* 4x10 5x10 6x10 4.5x10l HBP-1047- 7xl0' nt* 2.5xl0' 5xl0 2x10 5xl0 8x0 doxo It appears that, in vitro, the sensitivity of the cells to the peptide-doxorubicin conjugates is on the same order of magnitude as that of natural doxorubicin. The lines resistant to doxorubicin (KS62R and MCF7R) require a greater concentration of doxorubicin.
c) In vivo Evaluation of the Anti-tumor Activity of Peptide-Doxorubicin Coniugates Nude mice are injected with 3x10 6 HH9 cells subcutaneously in the side. On day 19 after the cell implant, the tumors are measured, and 4 groups of 6 mice are formed that receive by peritumoral injection either NaCI, or natural doxorubicin, or 1047-doxorubicin, or HBP1-doxorubicin conjugates at the rate of pg of doxorubicin or the equivalent of conjugates every two weeks. The tumors are measured and their volume calculated. On day 60, after injection of a total of 120 pg of doxorubicin or the equivalent, the tumor growth is inhibited by 77% with the 1047-doxorubicin conjugate, by 84% with the HBP1doxorubicin conjugate and by 79% with the doxorubicin alone, respectively, which shows that in vivo these conjugates are at least as effective as natural doxorubicin.
d) In vivo Evaluation of the Toxicity of Peptide-Doxorubicin Coniugates Injection of doxorubicin or doxorubicin-peptide by the intravenous route makes it possible to 0compare the toxicity of the different preparations. The estimate is made by the weight loss of the mice mice per group). Two experiments were done: Experiment 1: The mice received 200 ig per injection at 3-day intervals, or 600 Ag of doxorubicin or the equivalent peptide-doxorubicin in all. The mice were weighed the day after the last injection.
IO
Experiment 2: The mice received 2 injections of 200-pg of doxorubicin or the peptide-doxorubicin 00 equivalent at one week intervals, or 400 pg in all, and were weighed the day after the last injection.
Table 21 below reports the weight loss of the mice treated with doxorubicin or peptide-doxorubicin conjugates. The average weight loss is calculated in relation to the average weight of the mice that received C- NaCI.
STable 21 Groups NaCI Doxo. HBP3 HBP6 HBP7 HBP10 HBP13 Exp. 1 Weight 23.11 16.06 21.45 20.3 19.11 21.15 **nt Loss* 31* 8 12 18 9 Exp. 2 Weight 31.4 26.7 29.5 29.5 31.4 32.5 Loss* 15 6 6 0 0 4 not tested At the 600 pg dose, the mice that received the natural doxorubicin lost 31% of their weight, while those that received peptide-doxorubicin lost only 18% to 8% of their weight. The 400 pg-dose entailed a weight loss in mice that received natural doxorubicin, while the loss was not significant with the peptide-doxorubicin conjugates.
13) Transport of Ubiquitin Ubiquitin is a polypeptide of 76 amino acids, highly preserved during evolution, present in the organisms of all eukaryotes, with a molecular weight of 8500 Daltons. Intracellular ubiquitin is involved in various cell functions, such as breaking down proteins after their ligation with ubiquitin, the progression of the cell cycle, regulation of the activation of the transcription factor NF-kB. Extracellular ubiquitin has the property of inhibiting the proliferation of hematopoetic stem cells 0 by induction of apoptosis.
a) Coupling Ubiquitin with a Peptide.
1 mg of ubiquitin in 0.3 ml of sodium phosphate buffer 0.lm, pH 7 containing 0.15 M NaCI is added to 390 pg of SMCC (succinimidyl 4-(N-maleimidomethyl)cyclohexane-l-carboxylate) in 39 pl of I dimethylsulfoxide, and the solution is incubated for 30 minutes at laboratory temperature. The excess reagent is eliminated by centrifugation on ultrafiltration membranes (cutoff threshold 5,000 Dalton, Sartorius), followed by three washings in sodium phosphate buffer 0.1M, pH 7 containing 0.15M NaCI.
The peptide is then coupled in a molar ratio of 5 peptides to 1 ubiquitin in 1 ml of sodium phosphate buffer 0.1M, pH7, at laboratory temperature. Then, the excess uncoupled peptide is eliminated by centrifugation 0 on membranes, as in the preceding step.
b) Evaluation of Penetration of Peptide-Ubiquitin Conjugates Penetration of the peptide-ubiquitin conjugates is evaluated with conjugates prepared with 00 peptides with a biotin on the N-terminal side.
SHT29 cells are sown the night before in 24-well plates (5xl0 4 /wells).
SThe different (biotinylated) peptide-ubiquitin conjugates are added to decreasing concentrations t) (100 to 25 ;lg/ml) in the culture medium for 4 hours at 37°C. At the end of the culture, the cells are washed three times with PBS, then fixed 15 minutes in ethanol at -20 0 C. The penetration of the peptide-biotin- ,l ubiquitin conjugate is evaluated after incubation for 60 minutes with avidin coupled with peroxidase. The activity of the peroxidase is indicated by diaminobenzidine in the presence of H 2 0 2 [Figures and sequence listing appear here in text, but are added to end of translation Translator] The penetration, dependent on the concentration of the conjugates in the culture medium, is evaluated under a microscope by the intensity of the coloration expressed by crosses. Table 22 below gives the penetration of peptide-ubiquitin in the cells.
Table 22 Concentration (ag/ml) 100 50 Natural ubiquitin 1047-ubiquitin HBP7-ubuquitin very intense; intense; positive; slightly positive; at the limit of the reading.
c) Evaluation of Biological Activity of the Peptide-Ubiquitin Conjugates.
The proliferation of cells is measured by the test with MTT. Some Daudi (3xl0 4 cells/wells) were cultivated in 96-well plates for 48 hours at 37 0 C in the complete culture medium RPMI plus decreasing concentrations (200-12.5 ig/ml) of natural ubiquitin or ubiquitin conjugated with peptides, and the culture was continued for 48 hours.
Then, the wells are filled with 50 pl of MTT (3-,(4,5-dimethylthiazol-2yl)-2.5-diphenyl tetrazolium bromide) at 1 mg/ml in the culture medium and cultured for 4 hours.
0 The supernatant is removed, and 100 pl of dimethylsulfoxide is added. After the crystals dissolve, ci the coloration is read at 550 nm.
Table 23 shows the inhibition of the growth of Daudi cells by the peptide-ubiquitin conjugates.
The results, shown in Table 23, are calculated as a percentage of the average optical density obtained in the wells of cells containing the dilutions of the samples being tested and the optical density of the wells that O had only medium. They are expressed as a concentration giving 50% inhibition of growth of the cells.
Table 23 00 M Concentration (pg/ml) 200 100 Natural ubiquitin 0* 0 0 C1 1047-ubiquitin 60 21 0 0 HBP7-ubuquitin 96 68 16 11 0 of inhibition calculated in relation to the growth of cells that received only culture medium.
Natural ubiquitin does not inhibit the growth of Daudi cells. The conjugate prepared with peptide does not inhibit very much, while the conjugate prepared with peptide HBP7 is effective and still gives 35% inhibition at 50 pg/ml. The conjugate prepared with peptide 1047 gives intermediate inhibition values.
14) Transport ofCvtochrome C Cytochrome C is a hemoprotein that constitutes the mitochondrial lipid-protein complex. It plays a vital role in the cellular oxidation of plants and mammals. It is made up of a single polypeptide chain of 104 amino acids (molecular weight 13,000 Daltons) with the hth group attached to the cystein residues.
Graining of cytochrome C of the mitochondria in cytosol causes apoptosis of the cells by activating the caspases.
a) Coupling Cvtochrome C to Peptides To 4 mg of cytochrome C in 930 pl of sodium phosphate buffer 0.1M, pH7 containing 0.15M NaCI is added 540 pg of SMCC (succinimidyl 4-(N-maleimidomethyl)cyclohexane-l-carboxylate) in 54 pl of dimethylsulfoxide, and the solution is incubated 30 minutes at laboratory temperature. The excess reagent is eliminated by centrifugation on ultrafiltration membranes (cutoff threshold 10,000 Daltons, Sartorius), followed by three washings in sodium phosphate buffer 0.1M, pH7 containing 0.15M NaCI.
The peptide is then coupled in a molar ratio of 10 peptides to 1 cytochrome C in 1 ml of sodium phosphate buffer 0.1M, pH7 containing 0.15 M NaCI, for 3 hours at laboratory temperature. Then, the excess uncoupled peptide is eliminated by centrifugation/filtration as in the preceding step.
b) Evaluating the Penetration of the Peptide-Cvtochrome C Coniugates into the Cells t The penetration of the peptide-cytochrome C conjugates is evaluated with some conjugates Sprepared with peptides having a biotin on the N-terminal side.
H1299 cells are cultivated in the presence of the different (biotinylated) peptide-cytochrome C c conjugates at decreasing concentrations (100 to 25 jtg/ml) in the culture medium for 4 hours at 37 0 C. At the end of the culture, the cells are washed three times with PBS, then fixed 15 minutes in ethanol at -200. The O penetration of the peptide-biotin-cytochrome C conjugate is evaluated after incubation for 60 minutes with some avidin coupled with peroxidase. The cells are then washed with PBS, and the activity of the 00 peroxidase is indicated by diaminobenzidine in the presence of H 2 0 2 SThe penetration dependent on the concentration of conjugates in the culture medium is evaluated Sunder the microscope by the intensity of the coloration expressed by crosses. Table 24 indicates the Spenetration of the peptide-cytochrome C conjugates in the cells.
STable 24 Concentration (tg/ml) 100 50 Natural cytochrome C 1047-cytochrome C intense; positive; slightly positive; negative.
The table above shows that natural cytochrome C does not penetrate the cells and that the 1047cytochrome C conjugate penetrates effectively up to 25 pl/ml.
c) Evaluating the Biological Activity in vitro of Peptide-Cvtochrome C Conjugates H1299 or HT29 cells (3 x 104 cells/well) are cultivated in 96-well plates for 48 hours at 37°C in the complete culture medium RPMI with decreasing concentrations (200-3 Ig/ml) of natural cytochrome C or cytochrome C conjugated with peptides added, and the culture is continued for 48 hours.
The wells are then filled with 50 ul ofMTT (3-,(4,5-domethylthhiazol-2yl)-2,5-diphenyl tetrazolium bromide) at 1 mg/ml in the culture medium and are cultured for 4 hours.
The supernatant is removed, and 100 pl of dimethylsulfoxide is added to the wells. After the crystals dissolve, the coloration is read at 550 nm.
The results are calculated as a percentage of the mean optical density obtained in the wells of cells containing the dilutions of the samples being tested, and the optical density of the wells that received only medium. Table 25 gives the inhibition of growth of the cells by the peptide-ubiquitin conjugates.
Table On H1299 cells Concentration (g/ml) 100 50 25 12.5 6 3 -1 Natural cytochrome C 0* 0 0 0 0 0 0 1047-cytochrome C 46 40 29 15 14 0 On HT29 cells 00 Concentration (ug/ml) 200 100 50 25 12.5 0 Natural cytochrome C 12* 19 10 0 0 1047-cytochrome C 28 23 23 0 0 HBP3-cytochrome C 42 22 15 0 0 SHBP6-cytochrome C 41 18 12 2 0 Ni HBP7-cytochrome C 35 14 5 0 0 inhibition calculated in relation to the growth of cells that received only culture medium.
The table above shows the effectiveness of the peptide-cytochrome C conjugates on two types of cells. Natural cytochrome C does not inhibit the growth. At 50 pg/ml, the peptide most effective on HT29 cells is peptide 1047; peptides HBP3 and HBP6 give intermediate values, while peptide HBP7 is not very active at this concentration.
Transporting Anti-tumor and Anti-inflammatory Substances Using the Peptides in the Invention.
a) Transporting an Aspirin Derivative.
Analgesics like aspirin, long used in therapy, have been shown to have other effects, for example, the prevention of tumors of the colon in men and women who take aspirin regularly over a long period of time. In animals, the anti-cancer effect has been shown on various tumors. These effects have been partly attributed to their ability to inhibit the production of prostaglandins, by inhibiting the enzyme prostaglandin H synthetase and the cyclooxygenases.
Two aspirin derivatives conjugated to the peptide in sequence SEQ ID NO: 30: a salicylyl-HBP6 in -N terminal derivative (peptide SEQ ID NO: 47) and an HBP6-salicylyl in -C terminal derivative (SEQ ID NO: 48) have been tested for their ability to inhibit the growth of HT29 tumor cells in comparison with the natural peptide.
HT29 cells (3x10 4 cells/well) are cultured in 96-well plates for 48 hours at 37 0 C in the complete culture medium DMEM with decreasing concentrations (200-3 ug/ml) of aspirin or peptide-salicylyl conjugate added, and the culture continued for 48 hours.
The wells are then filled with 50 pl ofMTT (3-,(4,5-dimethylthiazol-2yl)-2,5-diphenyl tetrazolium bromide) at 1 mg/ml in the culture medium and cultured for 4 hours.
The supernatant is removed, and 100 pl of dimethylsulfoxide is added to the wells. After the crystals dissolve, the coloration is read at 550 nm.
The results inhibiting the growth of HT29 cells are shown in Table 26 and are calculated as a percentage of the mean optical density obtained in the wells of cells containing the dilutions of the samples being tested and the optical density of the wells that received only medium.
Table 26 Concentration (ug/ml) 1000 500 200 100 Natural aspirin 48* 6 3 0 Salicylyl-HBP6 51 35 30 HBP6-salicylyl 50 45 20 inhibition The addition of the peptide to the salicylic acid increases the aspirin's power to inhibit the growth of the cells.
b) Transporting a Phthalic Acid Derivative N-phthalimidoglutarimide (thalidomide) is a molecule that has a great variety of properties, such as a teratogenic effect, reduction of the production of TNF-a by the monocytes and suppression of angiogenesis. It has been shown that the teratogenic activity depends on the glutarimide residue, while the antiangiogenic effect depends on the phthaloyl group. We therefore prepared peptide SEQ ID NO: 46 derived from peptide SEQ ID NO: 26 (HBP3) 2 and added glycyl phthaloyl in N-terminal. The technique used to test the activity of phthaloyl-HBP3 is the same as the one previously described for aspirin.
The results of the inhibition of the growth of HT29 cells shown in Table 27 are calculated as a percentage of the mean optical density in the wells of cells containing dilutions of the samples being tested and the mean optical density of the wells of cells that received only medium.
Table 27 Concentration (tg/ml) 1000 500 200 100 Phthaloyl-HBP3 66 28 29 HBP3 2 0 0 It appears that the phthaloyl derivative inhibits the growth of HT29 cells.
16) Increasing the Activity of Anti-microbial Substances Using the Peptides in the Invention.
a) Lysozvme Lysozyme is one of the major constituents of granules of human polynuclear lymphocytes. It is x also found in secretions. It is a muraminidase. Lysozyme lyses and kills gram-positive microorganisms by modifying the peptidoglycans on their surface. As lysozyme does not easily penetrate the outer membrane of the bacteria, coupling lysozyme to peptides can help with their penetration.
Coupling Peptides to Lysozyme 0 8 mg of lysozyme in 2.7 ml of sodium phosphate buffer 0.1M, pH7 containing 0.15M NaCI is added to 1.8 mg of SMCC (succinimidyl 4-(N-maleimidomethyl cyclohexane-1-carboxylate) in 186 ml pl 00 [sic] dimethylsulfoxide, and the solution is incubated 30 minutes at laboratory temperature. The excess n reagent is eliminated by centrifugation on ultrafiltration membranes (cutoffthreshold 5,000 Daltons, Sartorius), followed by three washings in sodium phosphate buffer 0.1M, pH 7 containing 0.15M NaCl.
The peptide is then coupled in a molar ratio of 5 peptides to 1 lysozyme in 1 ml of sodium phosphate buffer 0.1M, pH7 containing 0.15M NaCI, for 3 hours at laboratory temperature. Then, the excess uncoupled peptide is eliminated by centrifugation on membranes, as in the preceding step.
Evaluating the Activity of Lvsozvme Coupled to Peptides.
The gram-positive bacterial strain Escherischia coli (ATCC25922) is cultured until the semilogarithmic phase in Luria-Bertani (LB) medium. The bacteria are collected by centrifugation and resuspended in 1% bacto-peptone medium at a concentration of 5x10' cfu/ml (colony forming unit/ml).
Lysozyme or the peptide-lysozyme conjugates are diluted half and half (256 to 0.5 jg/ml) in bacto-peptone medium, and 50 pl is distributed in 96-well plates. 50 jl of the bacterial dilution is then added to each well.
After 16 hours of incubation at 37 0 C, 10 pl from each well is smeared on gelose plates in LB medium.
After 18 hours of incubation at 37°C, the minimal bactericide concentration (MBC) is determined as the concentration of lysozyme or lysozyme-peptide that inhibits 75% of the growth.
Results Table 28 reports the inhibition of the growth ofE. coli (MBC pjg/ml).
Table 28 Natural lysozyme 256 1047-lysozyme 32 (HBPl) 3 -lysozyme 4 (HBP3) 2 -lysozyme 16 HBP6-lysozyme 16 The peptide-lysozyme conjugates have an antibacterial activity more effective than that of the natural lysozyme.
b) Antimicrobial Peptides V For several years, there has been considerable interest in studying the structure-function of short peptides having antibacterial and antifungal activities. Many natural peptides with varied sequences found in the animal and vegetable world have similar modes of action against a wide variety of microbes.
The antimicrobial peptides generally have an equivalent number of polar and non-polar residues within amphipatic areas and enough basic residues to give a peptide with an overall positive charge at DO neutral pH.
The peptides that are the subject of this invention are also mainly alkaline peptides, with a high affinity for heparin, and in the examples given below, it is shown that several of them have antibacterial 00 activity in vitro at concentrations at which they are not toxic to eukaryotic cells.
Evaluating the Antibacterial Activity of Cationic Peptides SThe gram-positive bacterial strains are: Enterococcusfaecalis (ATCC 29212), and Staphylococcus Vt aureus (ATCC 25923) and the gram-negative strains are: Escherichia.coli (ATCC 25922), Pseudomonas aeruginosa (ATCC 27853) and Salmonella typhimurim (clinical isolate), Salmonella typhiTy2 (WHO). The rl bacteria are cultured with shaking all night at 37 0 C, then recultured at a dilution of 1:50 in some fresh Luria-Bertani (LB) medium for 2 hours. The bacterial concentration is adjusted to 106 cfu/ml, and 50 pl is distributed in 96-well plates with an equal volume of peptides diluted half and half (256-0.5 pg/ml) in LB medium. After 16 hours at 37C, the minimum inhibiting concentration (MIC) is determined as the smallest concentration that totally inhibits the growth of the bacteria (total absence of turbidity). To determine the minimum bactericide concentration, 10 pl from each well is spread on gelose plates in LB medium. After 18 hours of incubation at 37C, the minimum bactericide concentration (MBC) is defined as the peptide concentration that allows only 0.01% of live bacteria to subsist.
Results.
The results are expressed as the peptide concentration in MBC (pM).
Table 29 Peptide strains E. coli S. typhimurium S. typhi-Ty2 P. aeruginosa S. aureus E. faecalis SEQ ID NO: 2 >160* >160 nt >160 >160 >160 SEQ ID NO: 3 13.25 13.25 1.65 26.5 >106 >106 SEQ ID NO: >170 >170 >170 >170 >170 >170 38 SEQ ID NO: 55 110 27.5 27.5 >110 >110 24 SEQ ID NO: 1.7 1.27 0.64 >110 >110 >110 44 SEQ ID NO: 1 1 0.66 0.5 >680 >680 Ampicillin 22 11 5.5 >696 <0.68 2.75 pM T The results in the above table show that peptide (HBP1)3 has bactericide activity, while peptides (HBPI)2 and HBPI3 are not active. However, when peptide HPB13 (SEQ ID NO: 38) is coupled with IN peptide SEQ ID NO: 24, it enhances its activity by a factor of 50 to 100 (peptide SEQ ID NO: 45). The same is true when peptide SEQ ID NO: 24 is coupled with peptide SEQ ID NO: 30 (HPB7) which itself has no anti-microbial activity.
00 M The minimum bactericide concentration (MBC) of all the peptides in the above table is equivalent to the MIC or differs by a dilution (MBC 2 x MIC). None these peptides are active on gram-positive Sbacteria.
SThe entire disclosure in the complete specification of our Australian Patent Application No. 40748/01 is by this cross-reference incorporated into the present specification.
REFERENCES
1) Cardin A.D. Weintraub H.J.R. Arteriosclerosis 9:21 (1989) 2) Merton B. et al. Annu. Rev. Cell Biol. 8:365 (1992) 3) David G. FASEB J. 7:1023 (1993) NO 4) Salmivira M Jalkanen M Experientia 51:863 (1995) Caldwell et al. Int. J. Biochem. Cell Biol. 28:203 (1996) 0 6) Fromm J.R. et al. Arch. Biochem. Bioph. 343:92 (1997) S7) Hirsch J. New Engl. J. Med. 324:1565 (1991) 8) Castellot J.J. et al. J. Cell Biol. 102:1979 (1986) 9) Ruoslahti E. Yamazuchi Y. Cell 64:867 (1991) l 10) Robinson D.S. Adv. Lip. Res. 1:1 (1963) S11) Kallunski P. Tryggvason K. J. Cell Biol. 116:559 (1992) 12) Cardin A.D. et al. Biochem. Biophys. Res. Cor. 154:741 (1988) 13) Avrameas A. et al. Proc. Natl. Acad. Sci. 95:5601 (1998) 14) Stevenson F.K. et al. J. Autoimmunity 6:809 (1993) Hirabayashi Y. et al. Scand. J. Immunol. 37:533 (1993) 16) Kalsi J.K. et al. Lupus 4:375 (1995) 17) Campanelli JT, Gayer GG and Scheller RH Development (1996) 122: 1663-1672 18) Fowlkes JL, Thrailkill KM, George-Nascimento Rosenberg CK Serra DM. Endocrinol (1997) 138: 2280-2285.
19) Maher DW, Lee BA Donoghue DJ Mol Cell Biol (1989) 9:2251-2253.
Inoue Watanabe Morino Tanaka Amachi T. Sasaki J. FEBS (1990) 269:89- 92.
21) Pasqualini Koivunen E. Ruoslahti E. Nature Biotech. (1997) 15:542-546.
22) Arkonac BM, Foster LC, Sibinga NES, Patterson C, Lai K, Tsai JC, Lee ME, Perrella MA Haber E. J Biol Chem (1998) 273: 4400-4405.
23) Yayon A, Klagsbrun M, Esko JD, Leder P. Omitz DM. Cell (1991) 64:841-848.
24) Gongqiao XU, Fornster GG Fornster JF Glyconjug J. (1996) 13:81-90.
Lortat-Jacob H Grimaud JA. FEBS (1991) 280: 152-154.
26) Hasan Najjam Gordon MY, Gibbs RV Rider CC. J Immunol (1999) 162: 1064- 1070.
27) Amara A, Lorthioir O, Valenzuela A, Magerus A, Thelen M, Montes M, Virelizier JL, Delepierre M, Baleux F, J. Biol. Chem. (1999) 272: 200-204.
28) Pohl J, Pereira HA, Martin NM Spitznagel JK. Amino acid sequence of CAP37 FEBS (1990) 272: 200-204.
29) Javadpour MM, Juban MM, Lo WCJ, Bishop SM, Iberty [sic] YB. Cowell SSM, Becher CL McLaughilin ML. J Med Chem (1996) 39: 3107-3113.
Ternynck T. and Avrameas S. "Techniques immunoenzymatiques" [Immunoenzymatic Techniques] Editions INSERM (1987).
00 List of Figures Fig. 1 pg ofstreptavidin-peroxidase for 10 4 cells Fig. 2 pg of peroxidase conjugate for 104 cells Fig. 5 pg ofIgG 1 for 10 4 cells 0 CHO cells human H1299 cells Fig. 6 1: Ribonuclease A (RNase A) 2: Coupling to peptide 10 (human Cyst.-HBP-CDR3) 00 (c 3: Coupling to peptide 10 (human HBP-CDR3-Cyst.) Fig. 7: 1 and 2: Coupling to peptide 16 (mouse P3CDR3) S3: Coupling to peptide 5 (mouse HBP-CDR3) S4: Coupling to peptide 10 (human HBP-CDR3) Ribonuclease A (RNase A) ,1 6: Peptide 16 (mouse P3CDR3) Fig. 8: Cellular variability RNase A-vector Concentration (ug/ml) Natural RNase A SEQ ID NO: SEQ ID NO: SEQ ID NO: 16 Fig. 9: Volume (mm 3 Number of days Fig. 11A:A375 Cells graphs DO=f (concentration of Ab in ug/ml) Fig. 11B:5F12 Cells Fig. 12: of bond/internalization Time (h) bond internalization Fig. 13A:A375 Cells °C of Ab (ug/ml) Fig. 13B:SKOV3 Cells °C of Ab (ug/ml) Fig. 14A:SKOV3 Cells 1 h °C of Ab Fig. 14B:SKOV3 Cells 4 h OC ofAb Fig. 16A, B, C: ID/g tissue tumor, liver, kidney, lungs, spleen, heart, muscle, bone, skin, stomach, sm. intestine, colon, (Nl skeleton, blood Fig. 16D: tumor, kidneys, spleen, muscle, skin, sm. intestine, skeleton Fig. 16E: [same as 16A, B, C above] IND Fig. 16F: [same as 16D above] 00 o",

Claims (43)

1. An amino acid sequence having the capacity to facilitate the penetration of a substance of interest inside cells and/or cell nuclei, characterized by the fact that it is capable of reacting in vivo with the aminoglycans. IN 2. An amino acid sequence having the ability to facilitate the penetration of a substance of interest inside cells and/or cell nuclei, characterized by the fact that it is capable of reacting with the aminoglycans, and by the fact that it comes from a protein of human origin. 00 C¢f 3. The amino acid sequence in Claim 1 or 2, characterized by the fact that it is capable of reacting with heparin, the chondroitine sulfates and their derivatives.
4. The amino acid sequence in any one of the preceding claims, characterized by the fact that it attaches IV) itself to a receptor on the cell membrane and crosses said cell membrane because of the receptor. The amino acid sequence in any one of the preceding claims, characterized by the fact that it is coded by the germinal line.
6. The amino acid sequence in any one of the preceding claims, characterized by the fact that it comes from a protein synthesized by a human cell.
7. The amino acid sequence in any one of the preceding claims, characterized by the fact that it comes from one of the proteins capable of reacting with the aminoglycans of the heparin or chondroitine sulfate type.
8. The amino acid sequence in any one of the preceding claims, characterized by the fact that it comes from human lipoprotein B.
9. The amino acid sequence in any one of the preceding claims, characterized by the fact that it has a number of basic amino acids at least equal to 3. The amino acid sequence in any one of the preceding claims, characterized by the fact that it is comprised of, or contains, at least one group of amino acids corresponding to one of the following formulas: a) (XBBBXXBX)n; b) (XBBXBX)n; c) (BBXmYBBXo)n; d) (XBBXXBX)n; or e) (BXBB)n; in which: B is a basic amino acid, X is a non-basic amino acid, Y is either a direct bond or 1 to amino acids, m is a whole number between 0 and 5, n is a whole number between 1 and 10, and o is a whole number between 0 and
11. The amino acid sequence in any one of the preceding claims, chosen from among the group including the following sequences: SEQ ID NO: 1, either in at least a dimer state, or (ii) in a monomer or polymer state coupled with another amino acid sequence: SEQ ID NO: 2; SEQ ID NO: 3; SEQ ID NO: SEQ ID NO: 7; SEQ ID NO: 9; SEQ ID NO: 10; SEQ ID NO: 13; SEQ ID NO: 16; SEQ ID NO: 19; SEQ ID NO: 20; SEQ ID NO: 21; SEQ ID NO: 23; SEQ ID NO: 25; SEQ ID NO: 26; SEQ ID NO: 30; SEQ ID NO: 33; SEQ ID NO: 34; SEQ ID NO: 35; SEQ ID NO: 36; SEQ ID NO: 37; SEQ ID NO: 38 and SEQ ID NO: 39.
12. The amino acid sequence in Claim 10, characterized by the fact that it includes at least two domains, Sone of them including an amino acid sequence with the formula: a) XBBBXXBX; b) XBBXBX; c) BBXmYBBXo; d) XBBXXBX; or e) BXBB; and the other of these fields having an amino acid sequence with the formula: a) XBBBXXBX; b) XBBXBX; c) BBXmYBBXo; d) XBBXXBX; e) BXBB; and f) an antibody fragment; in which: B is a basic amino acid, X is a non-basic amino acid, Y is either a direct bond or 1 to amino acids, m is a whole number between 0 and 5, o is a whole number between 0 and 00 13. The amino acid sequence in one of Claims 10 or 12, characterized by the fact that X is a hydrophobic C€ non-basic amino acid.
14. The amino acid sequence in any one of the preceding claims having less than 100 amino acids. ,1 15. The amino acid sequence in any one of the preceding claims having less than 50 amino acids.
16. The amino acid sequence in any one of the preceding claims having less than 25 amino acids.
17. The combination of the amino acid sequence in any one of Claims 1 to 16 with a substance of interest.
18. The use of the amino acid sequence in any one of Claims 1 to 15 to prepare a composition designed to transfer substances of interest into cells.
19. The use of the amino acid sequence in Claim 17 to prepare vectors. The vector for intracytoplasmic and/or intranuclear in vivo transfer, characterized by the fact that it is comprised of or includes at least one amino acid sequence reacting with the aminoglycans.
21. The vector for intracytoplasmic and/or intranuclear transfer characterized by the fact that it is comprised of or includes at least one amino acid sequence reacting with the aminoglycans, which comes from a human protein.
22. The vector in one of Claims 20 or 21, characterized by the fact that said amino acid sequence is according to any one of Claims 1 to 16.
23. The vector in any one of Claims 20 to 22, characterized by the fact that said amino acid sequence is coupled to at least one antibody fragment.
24. The vector is any one of Claims 20 to 23, characterized by the fact that said amino acid sequence is coupled to at least one polyreactive antibody fragment. The vector in any one of Claims 20 to 24, characterized by the fact that said amino acid sequence is coupled to all or part ofa hypervariable region of an antibody.
26. The vector in any one of Claims 20 to 25, characterized by the fact that said amino acid sequence is coupled to a fragment of the heavy chain of an antibody.
27. The vector in any one of Claims 23 to 26, characterized by the fact that said antibody is a human anti- DNA antibody.
28. The vector in any one of Claims 23 to 27, characterized by the fact that said antibody is an IgM.
29. The vector in any one of Claims 23 to 27, characterized by the fact that said antibody is an IgG. The vector in any one of Claims 20 to 29, characterized by the fact that said amino acid sequence is coupled to all or part of at least one CDR2 region of an antibody I 0 31. The vector in any one of Claims 20 to 29, characterized by the fact that said amino acid sequence is 0 coupled to all or part of at least one CDR3 region of an antibody.
32. The vector in Claim 31, characterized by the fact that region CDR3 is selected in the group consisting Cof RTT79, NE-1 and RT72.
33. The vector in any one of Claims 20 to 32, characterized by the fact that said amino acid sequence is coupled to at least one substance of interest that can be incorporated naturally or non-naturally into cells and/or the nuclei of said cells.
34. The vector in Claim 33, characterized by the fact that its capacity to penetrate inside the cells is Cc substantially independent of the nature of said substance of interest. The vector in one of Claims 33 or 34, characterized by the fact that said substance of interest is coupled at N or C terminal end of the amino acid sequence. t 36. The vector in any one of Claims 23 to 35, characterized by the fact that the coupling is chemical.
37. The vector any one of the Claims 22 to 36, characterized by the fact that the coupling is done with homofunctional or heterofunctional bridge reagents of the type succinimidyl 4-(N- maleimidomethyl)cyclohexane-1-carboxylate (SMCC).
38. The vector in one of the Claims 22 to 35, characterized by the fact that the coupling is done by genetic engineering.
39. Thevector in any one of Claims 32 to 38, characterized by the fact that said substance of interest is chosen from the group including: nucleic acid, a protein, a drug, an antigen, an antibody. The vector in any one of Claims 33 to 39, characterized by the fact that it makes it possible to enhance the biological activity of the substance of interest to which it is coupled.
41. The vector in any one of Claims 33 to 40, characterized by the fact that it makes it possible to reduce the toxicity of the substance of interest to which it is coupled.
42. The vector in any one of Claims 33 to 41, characterized by the fact that it makes in vitro transfection of cells possible.
43. The vector in any one of Claims 20 to 42, characterized by the fact that it is coupled to at least one substance of interest with at least one anchoring molecule having a strong natural affinity for said substance of interest.
44. The vector in Claim 43, characterized by the fact that the anchoring molecule is chosen from among protein A, protein G, anti-IgG fragment F(ab') 2 anti-peroxidase IgG, anti-RNase F(ab') 2 concanavalin A, streptavidin and avidin. The vector in Claim 43 or 44, characterized by the fact that said anchoring molecule has an affinity for a large number of substances of interest.
46. An eukaryotic cell, characterized by the fact that it contains an amino acid sequence in any one of Claims 1 to 16.
47. An eukaryotic cell, characterized by the fact that it contains a vector in any one of Claims 20 to
48. An eukaryotic cell transfected by means of a vector in one of Claims 20 to S49. A process for in vitro transfer of a substance of interest inside a cell, characterized by the fact that it includes the following steps: a) coupling said substance of interest to an amino acid sequence in one of Claims 1 to 16, or to a vector in one of Claims 20 to 45, and b) incubating the cell with said coupling product at a culture temperature that permits active metabolism of said cell. The process in Claim 49, characterized by the fact that the incubation in step b) is done at a temperature between 25°C and 39°C. 00
51. The process in one of Claims 49 or 50, characterized by the fact that the incubation in Step b) is done at 37 0 C.
52. A biological, pharmaceutical, cosmetic, agro-food, diagnostic or tracking composition, characterized (-i t by the fact that it has as its active ingredient either the vector in one of Claims 20 to 45, or cells in one of Claims 46 to 48.
53. The use of a composition in Claim 52 for the formulation and preparation of biological, pharmaceutical, cosmetic and agro-food, diagnostic or tracking products.
54. A diagnostic agent characterized by the fact that it has at least one vector in one of Claims 19 to and/or one cell in Claims 46 to 48. A diagnostic kit, characterized by the fact that it includes, in one or more containers, a predetermined quantity of the composition in Claim 54.
56. The vector for intracytoplasmic transfer in vivo in Claim 20, characterized by the fact that it is composed of, or includes, at least one amino acid sequence having between 10 and 21 amino acids, in which at least 20% of all amino acid residues are lysine residues and at least 50% of all amino acid residues are basic amino acid residues.
57. The vector in Claim 56, characterized by the fact that it is composed of, or includes, at least one amino acid sequence chosen from among sequences (HB1)3, HBP6, (HBP3) 2 HBP7, HBP11, HBP 13 and HBP2.
58. A vector for intranuclear transfer in vivo in Claim 20, characterized by the fact that it is composed of, or includes, at least one amino acid sequence having at least 14 amino acids, in which at least 35% of all residues are basic amino acid residues and contain no lysine residues.
59. The vector in Claim 58, characterized by the fact that it is composed of, or includes, at least one amino acid sequence chosen from among sequences HBP10 and The vector in any one of Claims 56 to 59, characterized by the fact that it is conjugated with a biologically active substance. Dated this 6th day of May 2005 DIATOS S.A. By their Patent Attorneys GRIFFITH HACK Fellows Institute of Patent and Trade Mark Attorneys of Australia 62 SEQUENCE LISTING <11I0> DIATOS S.A. <120> AMINO ACID SEQUENCES FACILITATING THE PENETRATION OF A SUBSTANCE OF INTEREST INSIDE CELLS AND/OR CELL NUCLEI <130> 12052-PCT-DIATOS <140> <141> <1 50> <151> PCT/FROO/0262 1 200 1-03-01 FROO/0262 I
2000-03-01 <160> 48 <170> Patent in version <210> <21 1> <212> <213> 1 7 PRT Honmo sapiens <400> 1 Val Lys Arg Gly Leu Lys Leu 1 <2 10> <211 <212> <213> 2 14 PRT Homo sapiens <400> 2 Val Lys Arg Gly Leu Lys Leu Val Lys Arg Gly Leu Lys Leu <210> 3 <21 1> 21 <212> PRT <213> Homo sapiens <400> 3 Val Lys Arg Gly Leu Lys Leu Val Lys Arg Gly Leu Lys Leu Vat Lys 1 5 10 Arg Gly Leu Lys Leu <210> <21 1> <212> <213> 4 11 PRT Mus musculus x Rattus norvegicus <400> 4 Arg Gin Lys Tyr Asn Lys Arg Ala Met Asp Tyr <210> <21 1> <212> <213> 18 PRT Homo sapiens-Mus musculus-Rattus norvegicus <400> Val Lys Arg Gly Leu Lys Leu Arg Gin Lys Tyr Asn Lys Arg Ala Met 1 5 10 Asp Tyr <210O> 6 <21 1> <212> PRT <213> Mus musculus x Rattus norvegicus <400> 6 Thr Tyr Tyr Ser Asp Thr Val Lys Gly Arg Phe Thr Arg Gin Lys Tyr 1 5 10 Asn Lys Arg Ala <210> 7 <211I> 27 <212> PRT <213> Homo sapiens-Mus musculus-Rattus norvegicus <400> 7 Val Lys Arg Gly Leu Lys Leu Thr Tyr Tyr Ser Asp Thr Val Lys Gly 1 5 10 Arg Phe Thr Arg Gin Lys Tyr Asn Lys Arg Ala <210> <211I> <212> <213> 8 17 PRT Homo sapiens <400> 8 Val Arg Arg Ser Gly Arg Val Val Val Pro Ala Ala Pro Arg Asn Arg 1 5 10 Asp <210> 9 <211> 24 <212> PRT <213> Homo sapiens <400> 9 Val Lys Arg Gly Leu Lys Leu Val Arg Arg Ser Gly Arg Val Val Val 1 5 10 Pro Ala Ala Pro Arg Asn Arg Asp <210> <211> 19 <212> PRT <213> Homo sapiens <400> Val Lys Arg Gly Leu Lys Leu Arg His Val Arg Pro Arg Val Thr Arg 1 5 10 Met Asp Val <210> 11 <211> 19 <212> PRT <213> Homo sapiens <400> 11 Val Lys Arg Gly Leu Lys Leu Gly Tyr Tyr Asp Phe Trp Ser Gly Pro 1 5 10 Gly Lys Asn <210> 12 <211> 27 <212> PRT <213> Mus musculus x Rattus norvegicus <400> 12 Asn Val Lys Lys Pro Lys Leu Thr Tyr Tyr Ser Asp Thr Val Lys Gly 1 5 10 Arg Phe Thr Arg Gin Lys Tyr Asn Lys Arg Ala <210> 13 <211> 31 <212> PRT <213> Homo sapiens <400> 13 Val Lys Arg Gly Leu Lys Leu Ser Gly Ser Thr Asn Tyr Asn Pro Ser 1 5 10 Leu Lys Ser Arg His Val Arg Pro Arg Val Thr Arg Met Asp Val 25 <210> <211> <212> <213> 14 28 PRT Mus musculus x Rattus norvegicus <400> 14 Arg Gin Lys Tyr Asn Lys Arg Ala Thr Tyr Tyr Ser Asp Thr Val Lys 1 5 10 Gly Arg Phe Thr Arg Gin Lys Tyr Asn Lys Arg Ala <210> <211> 24 <212> PRT <213> Homo sapiens <400> Arg His Val Arg Pro Arg Val Thr Arg Met Asp Val Arg His Val Arg 1 5 10 Pro Arg Val Thr Arg Met Asp Val <210> <211> <212> <213> 16 31 PRT Homo sapiens <400> 16 Arg His Val Arg Pro Arg Val Thr Arg Met Asp Val Val Lys Arg Gly 1 5 10 Leu Lys Leu Arg His Val Arg Pro Arg Val Thr Arg Met Asp Val 25 <210> 17 <211> 8 <212> PRT <213> Homo sapiens <400> 17 Leu Arg Lys Arg Leu Leu Arg Asp <210> 18 <211> 4 <212> PRT <213> Homo sapiens <400> 18 Lys Ser Arg Lys 1 <210O> 19 <21 1> 8 <212> PRT <213> Homo sapiens <400> 19 Lys Ser Arg Lys Lys Ser Arg Lys 1 <210> <21 1> 12 <212> PRT <213> Homo sapiens <400> Arg Lys Gly Phe Tyr Lys Arg Lys Gin Cys Lys Pro 1 5 <210> 21 <21 1> 24 <212> PRT <213> Homo sapiens <400> 21 Gly Arg Pro Arg Glu Ser Gly Lys Lys Mrg Lys Arg Lys Arg Leu Lys <210> <21 1> <212> <213> 22 32 PRT Mus musculus x Rattus norvegicus <400> 22 Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Thr Tyr Tyr Ser Asp Thr Val Lys Gly Arg Phe Thr Arg Gin Lys Tyr Asn Lys Arg Ala 25 <210> 23 <21 1> <212> PRT <213> Mus musculus x Rattus norvegicus <400> 23 Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Val Lys Arg Gly Leu Lys Leu Arg Gin Lys Tyr Asn Lys Arg Ala Met Asp Tyr 25 <210O> 24 <21 1> <212> <213> 14 PRT Synthetic peptide <400> 24 Lys Leu Ala Lys Leu Ala Lys Lys Leu Ala Lys Leu Ala Lys <210> <211> 18 <212> PRT <213> Homo sapiens <400> Arg Lys Gly Lys Phe Tyr Lys Arg Lys Gin Cys Lys Pro Ser Arg Gly 1 5 10 Arg Lys <210> 26 <211> 18 <212> PRT <213> Homo sapiens <400> 26 Ser Glu 1 Glu Ser <210> <211> <212> <213> Arg Lys Lys Arg Arg Arg Glu Ser Arg Lys Lys Arg Arg Arg 5 10 27 18 PRT Homo sapiens <220> <221> miscfeature <222> <223> All amino acids are in configuration D <400> 27 Ser Glu Arg Lys Lys Arg Arg Arg Glu Ser Arg Lys Lys Arg Arg Arg 1 5 10 Glu Ser <210> 28 <211> 16 <212> PRT <213> Homo sapiens <400> 28 Arg Lys Lys Arg Arg Arg Gly Asp Mrg Lys Lys Arg Arg Arg Gly Asp 1 5 10 <210> 29 <211I> <212> PRT <213> Homo sapiens <400> 29 Val Lys Arg Gly Lett Lys Lett Leut Arg Lys Arg Leut Leut Arg Asp 1 5 10 <210> <211> <212> <213> PRT Homo sapiens <400> Gly Lys Arg Lys Lys Lys Gly Lys Leut Gly Lys Lys Arg Asp Pro 1 5 10 31 <211I> 16 <212> PRT <213> Homo sapiens <220> <221> misc feature <222> 0..0 <223> The amino acids are in front-back position. <400> 31 Pro Asp Arg Lys Lys Gly Leu Lys Gly Lys Lys Lys Arg Lys Lys Gly 1 5 10 <210> 32 <211> 14 <212> PRT <213> Homo sapiens <220> <221> misc feature <222> <223> All amino acids are in configuration D. <400> 32 Gly Lys Arg Lys Lys Lys Gly Lys Leu Gly Lys Lys Arg Asp 1 5 <210> 33 <211> 13 <212> PRT <213> Homo sapiens <400> 33 Gly Leu Lys Lys Asn Gly Ser Cys Lys Arg Gly Pro Arg <210> 34 <21 1> <212> PRT <213> Homno sapiens <400> 34 Tyr Arg Ser Arg Lys Tyr Ser Ser Trp Tyr Tyr Ala Leu Lys Arg 1 5 10 <210> <211I> <212> <213> 14 PRT Homio sapiens <400> Ser Arg Arg Ala Arg Arg Ser Pro Arg His Leu Gly Ser Gly 1 5 <210> 36 <211I> <212> <213> PRT Homo sapiens <400> 36 Ala Lys Thr Gly Lys Arg Lys Arg Ser Gly 1 5 <210> 37 <211I> 13 <212> PRT <213> Homo sapiens <400> 37 Gin Gly Lys Ser Lys Arg Glu Lys Lys Asp Arg Val Phe 1 5 <210> <211I> <212> <213> 38 12 PRT Homo sapiens <400> 38 Lys His Leu Lys Lys His Leu Lys Lys His Leu Lys 1 5 <210> <211I> <212> <213> 39 17 PRT Homo sapiens <400> 39 Leu Arg Arg Glu Arg Gin Ser Arg Leu Mrg Arg Glu Arg Ser Gin Ser 1 5 10 Arg <210> <211> <212> <213> 32 PRT Homo sapiens <400> Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Val Lys Arg 1 5 10 Gly Leu Lys Leu Arg His Val Arg Pro Arg Val Thr Arg Met Asp Val O 20 25 <210> 41 <211> 31 I <212> PRT O <213> Homo sapiens 00 <400> 41 Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Ser Glu Arg 1 5 10 Lys Lys Arg Arg Arg Glu Ser Arg Lys Lys Arg Arg Arg Glu Ser 25 <210> 42 <211> 27 <212> PRT <213> Homo sapiens <400> 42 Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Lys Ser Arg Arg 1 5 10 Ala Arg Arg Ser Pro Arg His Leu Gly Ser Gly <210> 43 <211> 21 <212> PRT <213> Synthetic peptide <400> 43 Val Lys Arg Gly Leu Lys Leu Lys Leu Ala Lys Leu Ala Lys Lys Leu Ala Lys Leu Ala Lys <210> 44 <211> <212> <213> 29 PRT Synthetic peptide <400> 44 Lys Leu Ala Lys Leu Ala Lys Lys Leu Ala Lys Leu Ala Lys Gly Lys 1 5 10 Arg Lys Lys Lys Gly Lys Leu Gly Lys Lys Arg Asp Pro <210> <211> 26 <212> PRT <213> Synthetic peptide <400> Lys Leu Ala Lys Leu Ala Lys Lys Leu Ala Lys Leu Ala Lys Lys His 1 5 10 Leu Lys Lys His Leu Lys Lys His Leu Lys <210> <211> <212> <213> <220> <221> 46 18 PRT Homo sapiens miscfeature <222> 0..0 <223> Residue ofglycine-phthaloyl in N-terminal position <400> 46 Xaa Gly Arg Lys Lys Arg Arg Arg Glu Ser Arg Lys Lys Arg Arg Arg 1 5 10 Glu Ser <210> 47 <211> <212> <213> <220> <221> <222> <223> 18 PRT Homo sapiens misc feature 0..0 Has a salicylic motif (named X) in N-terminal position. <400> 47 Xaa Gly Arg Pro Arg Glu Ser Gly Lys Lys Arg Lys Arg Lys Arg Leu 1 5 10 Lys Pro <210> 48 <211> 19 <212> PRT <213> Homo sapiens <220> <221> <222> <223> miscfeature 0..0 Has a salicylic motif (named X) in C-terminal position. <400> 48 Gly Arg Pro Arg Glu Ser Gly Lys Lys Arg Lys Arg Lys Arg Leu Lys 1 5 10 Xaa Pro Gly 0
AU2005201938A 2000-03-01 2005-05-06 Amino acid sequences facilitating penetration of a substance of interest into cells and/or cell nuclei Abandoned AU2005201938A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
FR00/02621 2000-03-01
AU40748/01A AU780785B2 (en) 2000-03-01 2001-03-01 Amino acid sequences facilitating penetration of a substance of interest into cells and/or cell nuclei
PCT/FR2001/000613 WO2001064738A2 (en) 2000-03-01 2001-03-01 Heparin-binding amino acid sequences derived from human proteins facilitating penetration of a substance of interest into cells and/or cell nuclei

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU40748/01A Division AU780785B2 (en) 2000-03-01 2001-03-01 Amino acid sequences facilitating penetration of a substance of interest into cells and/or cell nuclei

Publications (1)

Publication Number Publication Date
AU2005201938A1 true AU2005201938A1 (en) 2005-06-02

Family

ID=34596352

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2005201938A Abandoned AU2005201938A1 (en) 2000-03-01 2005-05-06 Amino acid sequences facilitating penetration of a substance of interest into cells and/or cell nuclei

Country Status (1)

Country Link
AU (1) AU2005201938A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114075544A (en) * 2020-08-21 2022-02-22 复旦大学附属华山医院 Method for preparing de-ubiquitin erythrocyte product based on ultrafiltration centrifugation

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114075544A (en) * 2020-08-21 2022-02-22 复旦大学附属华山医院 Method for preparing de-ubiquitin erythrocyte product based on ultrafiltration centrifugation

Similar Documents

Publication Publication Date Title
AU780785B2 (en) Amino acid sequences facilitating penetration of a substance of interest into cells and/or cell nuclei
AU2020200975B2 (en) New stable antibody-drug conjugate, preparation method therefor, and use thereof
USRE46211E1 (en) Delivery system using MAB 3E10 and mutants and/or functional fragments thereof
AU2011244107B2 (en) Peptide derivatives, preparation thereof and uses thereof as vectors
US8097704B2 (en) Antibody specifically binding to DR5 and composition for preventing or treating cancers comprising the same
US8119601B2 (en) Voltage dependent anion channel (VDAC1) compositions and methods of use thereof for regulating apoptosis
US8003595B2 (en) Amino acid sequences facilitating penetration of a substance of interest into cells and/or cell nuclei
KR20120125455A (en) Intracelluar targeting bipodal peptide binder
JP2015533838A (en) rTRAIL mutant and its monomethyl auristatin E conjugate
CN113891731A (en) Drug conjugates and methods of use thereof
US20120165269A1 (en) Tumor Cell-Killing Peptides
CN111093718A (en) Therapeutic nanoconjugates and uses thereof
US8648045B2 (en) VDAC1 compositions and methods of use thereof for regulating apoptosis
KR102166549B1 (en) A Blood-Brain Barrier Penetrating Peptide, and the Conjugate Comprising The Same
AU2005201938A1 (en) Amino acid sequences facilitating penetration of a substance of interest into cells and/or cell nuclei
CA2572825A1 (en) Compositions for multi-step delivery of hot-spots radiation to cancer cells
WO2022001710A1 (en) Intermediate for preparing antibody-drug conjugate (adc), preparation method therefor, and use thereof
Polli et al. Cell penetrating peptides conjugated to anti-carcinoembryonic antigen “catch-and-release” monoclonal antibodies alter plasma and tissue pharmacokinetics in colorectal cancer xenograft mice
CA3172475A1 (en) Ngr conjugates and uses thereof
WO2024026707A1 (en) Chimeric antigen receptor systems, methods of preparation, and uses thereof
CN113597318A (en) Therapeutic nanoconjugates and uses thereof
CN115845078A (en) Phase separation based tumor targeted drug delivery system
IL185500A (en) Vdac1 compositions and use thereof for preparation of medicaments for inducing apoptosis

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application