WO2010108262A1 - TISSUE KALLIKREIN FOR THE TREATMENT OF PANCREATIC β-CELL DYSFUNCTION - Google Patents

TISSUE KALLIKREIN FOR THE TREATMENT OF PANCREATIC β-CELL DYSFUNCTION Download PDF

Info

Publication number
WO2010108262A1
WO2010108262A1 PCT/CA2010/000413 CA2010000413W WO2010108262A1 WO 2010108262 A1 WO2010108262 A1 WO 2010108262A1 CA 2010000413 W CA2010000413 W CA 2010000413W WO 2010108262 A1 WO2010108262 A1 WO 2010108262A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
isolated
purified
klkl
cell
Prior art date
Application number
PCT/CA2010/000413
Other languages
French (fr)
Inventor
Mark Williams
Kevin Richardson
Original Assignee
Diamedica Inc.
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
Application filed by Diamedica Inc. filed Critical Diamedica Inc.
Priority to NZ595364A priority Critical patent/NZ595364A/en
Priority to EP10755352A priority patent/EP2411042A4/en
Priority to AU2010228068A priority patent/AU2010228068A1/en
Priority to JP2012501089A priority patent/JP2012521366A/en
Priority to CA2756801A priority patent/CA2756801A1/en
Priority to CN2010800181825A priority patent/CN102438648A/en
Publication of WO2010108262A1 publication Critical patent/WO2010108262A1/en
Priority to US13/241,882 priority patent/US20120070425A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • A61K38/4853Kallikrein (3.4.21.34 or 3.4.21.35)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Abstract

This invention relates to methods for treating pancreatic islet β-cell dysfunction and the conditions associated with pancreatic islet β-cell dysfunction, including administering a therapeutically effective amount of tissue kallikrein, variants or active fragments thereof.

Description

TISSUE KALLIKREIN FOR THE TREATMENT OF PANCREATIC P-CELL
DYSFUNCTION
CROSS REFERENCE TO RELATED APPLICATIONS This application claims the benefit of and priority to United States Patent
Application Nos, 61/163,173 filed March 25, 2009, under the title TISSUE
KALLIKREIN FOR THE TREATMENT OF PANCREATIC β-CELL DYSFUNCTION,
The content of the above patent applications is hereby expressly incorporated by reference into the detailed description hereof.
FIELD OF THE INVENTION
The present invention relates to methods of the treatment of pancreatic β-cell dysfunction and treating pancreatic diseases and conditions associated therewith by the modulation β-cells,
BACKGROUND OF THE INVENTION Both type I and type II diabetes mellitus are of great concern today. Approximately 24 million people in the United States are affected by the disease (Mueller, Phys Ther, 2008, 88(1 1): 1250-3) and the incidence is on the rise around the globe. While type I diabetes can only be treated by insulin injections, type II diabetes may be treated through diet and exercise in some cases. Diet and exercise can even ward off type Il diabetes development; however the increasing sedentary lifestyle in many regions of the world is resulting in obesity at epidemic proportions. For those who require it, treatment can be expensive and inconvenient, and may produce several undesirable side effects with the currently available drugs. β-cells, found in the pancreas, are responsible for the production and release of insulin into the blood stream. They represent the majority of the endocrine cells and form the core of the islets. The pancreatic β-cells secrete insulin in response to increasing glucose levels. Insulin aids in the entry of glucose into the muscle and fat cells.
(Ellingsgaard et al, PNAS, 2008, 105(35); 13162-7), In individuals with type I diabetes, the β-cells are attacked by an autoimmune response. The β-cells that remain are insufficient to produce enough insulin to remove the glucose from the blood. They show increased levels of β-cell destruction. For those with type II diabetes, the muscle and liver cells are no longer able to respond to normal blood insulin levels. Therefore they also end up with high blood glucose levels. This can result in β-cell death and loss of β- cell function as compared to healthy individuals. Long standing cases of type 1 diabetes show -99% deficiency in β-cell mass while cases of type II diabetes show -65% deficiency in β-cell mass within the pancreas (Meier, Diabetologia, 2008, 51 :703-13), Modulation of the levels of healthy β-cells, in particular, increasing the levels or activity of such cells, may therefore serve as an effective therapy to reverse and possibly prevent diabetes.
Modulation of β-cell levels has been attempted through the use of stem cells and organ transplant. These methods are seen to have some drawbacks (Meier, Diabetologia, 2008, 51 :703-13). Issues regarding stem cell supply and ethics may limit the ability of this therapy. Transplant presents the risks associated with any organ transplant, be it rejection, infection and/or subsequent mortality.
A therapy is therefore desired which can stimulate β-cell production thereby increasing the β-cell mass. This strategy will effectively serve to suppress glucagon secretion (Ellingsgaard et a!, PNAS, 2008, 105(35): 13162-7) and restore insulin production and secretion to normal levels, resulting in further suppression of glucagon secretion and hepatic glucose production, leading to an improvement in overall peripheral insulin action (Meier, Diabetologia, 2008, 51 : 703- 13).
Until the recent work of the present inventors, no one has contemplated the use of tissue kallikrein (KLKl) for the treatment of pancreatic islet β-cell dysfunction and treating diseases and conditions associated therewith by the modulation β-cell mass,
KLKl is a serine protease which cleaves low-molecular- weight kininogen resulting in the release of kallidin (lysl-bradykiπin), KLKl may be formulated to produce a product which can be delivered to modulate β-cell mass without any of the issues associated with the alternate proposed therapies (stem cell usage and pancreatic organ transplant). SUMMARY OF THE INVENTION
The present invention includes methods of the treatment of pancreatic islet β-cell dysfunction and treating diseases and conditions associated therewith by the modulation β-cell mass comprising administering a therapeutically effective dose of KLKl, variants of KLKl, or active fragments thereof.
In one aspect of the present invention the KLKl can be a purified/isolated natural form, a synthetic form or a recombinant form.
In another aspect of the present invention the isolated KLKl can be human KLKl (SEQ lD NO. 1) In another aspect of the present invention the isolated KLKl can be haiπadryas baboon KLKl (SEQ ID NO, 2)
In another aspect of the present invention the isolated KLKl can be crab eating macaque KLKl (SEQ ID NO. 3)
In another aspect of the present invention the isolated KLKl can be cotton top tamarin KLK 1 (SEQ ID NO, 4)
In another aspect of the present invention the isolated KLKl can be dog KLKI (SEQ ID NO. 5).
In another aspect of the present invention the isolated KLKl can be sheep KLKl (SEQ ID NO, 6), In another aspect of the present invention the isolated KLKl can be rabbit KLKl
(SEQ ID NO. 7).
In another aspect of the present invention the isolated KLKl can be bovine KLKl (SEQ ID NO, 8),
In another aspect of the present invention the isolated KLKl can be horse KLKl (SEQ ID NO, 9).
In preferred aspect of the present invention the isolated KLKl can be pig KLKl (SEQ ID NO, 10),
In addition, the present invention further provides pharmaceutical compositions and method of treating disease associated with reduced pancreatic islet β-cell functioning and/or reduced pancreatic islet β-cell mass. In a preferred embodiment the disease or condition associated with reduced pancreatic islet β-cell functioning and/or reduced pancreatic islet β-cell mass is type 1 or type II diabetes,
In a further aspect of the invention, modulation of β-cell mass can be an increase in the β-cell mass as compared to the diseased state,
In a further aspect of the invention modulation of β-cell mass can be β-cell regeneration.
In the present invention β-cell regeneration refers to the restoration of normal β- cell function by increasing the number of functional β-cell s or by fixing impaired β-cell by restoring normal function,
In yet a further aspect of the present invention modulation of β-cell mass can be an increase in β-cell proliferation.
In another aspect of the present invention, KLKl , or a variant or an active fragment thereof, can be administered orally. Oral administration may be an enteral administration, such as a liquid, pill, or capsule to be swallowed,
In a further aspect of the present invention, an oral therapeutic dose can be a maximum dose range of about 1 to about 1000 International Units (IU) per day.
Another aspect of the present invention includes a method as herein described further comprising the use of an additional therapeutic method useful in the modulation β-cell mass or treating diseases or conditions associated therewith. An additional therapeutic method includes, but is not limited to, stem cell transplant and pancreatic organ transplant.
Another aspect of the present invention includes a composition formulated for oral administration comprising about 1 to about 1000 IU of KLKl, or a variant or an active fragment thereof, optionally further comprising a pharmaceutically acceptable excipient, and optionally further comprising an additional therapeutic compound as described above. DETAILED DESCRIPTION Definitions
Tissue kallikreins are a family of serine proteases that are primarily noted for their role in controlling hypertension through cleavage of kininogen into lysyl-bradykinin
(kallidin) (Yousef et al., Endocrine Rev. 2001, 22: 184-204), As there are a large number of known human and animal tissue kallikreins, only one has kininogenase activity, i.e., the ability to release kinin, In humans this enzyme is known as KLKl or pancreatic/renal kallikrein. The inventors believe that KLK 1 appears to be a ubiquitous or multiple target acting enzyme, in addition to its recognized role in hypertension regulation and as such may specifically play an important role in the treatment of pancreatic islet cell dysfunction and treating diseases and conditions associated therewith by the modulation of β-cell mass. As used herein, the term "human tissue kallikrein" or KLKl is synonymous with the following terms: cal Herein, glumorin, padreatin, padutin, kallidinogenase, bradykininogenase, pancreatic kallikrein, onokrein P, dϊlmϊnal D, depot- Padutin, urokallikrein, or urinary kallikrein. Tissue kallikrein with similar kininogenase activity can also be found in various animals and therefore could be used in the treatment of pancreatic islet cell dysfunction,
A preferred embodiment of the present invention can be human tissue kallikrein precursor polypeptide (kidney /pancreas/salivary gland kallikrein) (KLKl) and has the following sequence (SEQ ID NO: 1):
NP_0G2248 GI:4504875 Homo sapiens KLKl_human
1-18 signal peptide 19-24 propeptide 25-262 mature peptide >gi|45G4875|ref]NP_0G2248, 1 1 kallikrein 1 preproprotein [Homo sapiens]
MWFLVLCLALSLGGTGAAPPIQSRIVGGWECEQHSQPWQAALYHFSTFQCGGIL
VHRQWVLTAAHCISDNYQLWLGRHNLFDDENTAQFVHVSESFPHPGFNMSLLE NHTRQADEDYSHDLMLLRLTEPADTITDA VK VVELPTEEPEVGSTCLASGWGSIE PENFSFPDDLQCVDLKILPNDECKKAHVQKVTDFMLCVGHLEGGKDTCVGDSG GPLMCDGVLQGVTSWGY VPCGTPNKPSVA VR VLS Y VKWIEDTIAEN S (SEQ ID NO:1) A further embodiment of the present invention includes hamadryas baboon tissue kallikrein (kidney/pancreas/salivary gland kallikrein) (SEQ ID NO. 2) which has 90% sequence identity to human KLK 1 (SEQ ID NO, 1 )
Q2877 KLK i_P APHA
MWFL VLCLALSLGGTGAAPPIQSRIVGGWECSQPWQAALYHFSTFQCGGIL VHP QWVLTAAHCIGDNYQLWLGRHNLFDDEDTAQFVHVSESFPHPCFNMSLLKNHT RQADEDYSHDLMLLRLTQPAEITDAVQVVELPTQEPEVGSTCLASGWGSIEPENF SYPDDLQCVDLKILPNDKCAKAHTQKVTEFMLCAGHLEGGKDTCVGDSGGPLT
CDGVLQGVTSWGYIPCGSPMKPAVFVRVLSYVKWIEDTIAENS (SEQ ID NO. 2)
A further embodiment of the present invention includes crab eating macaque tissue kallikrein (kidney/pancreas/salivary gland kallikrein) (SEQ ID NO. 3) which has 90% sequence identity to human KLKl (SEQ ID NO. 1):
Q07276-1 KLK1_MACFA NfWFLVLCLALSLGGTGRAPPIQSRIVGGWECSQPWQAAL YHFSTFQCGGIL VHP
QWVLTAAHCISDNYQLWLGRHNLFDDEDTAQFVHVSESFPHPGFNMSLLKNHT RQADDYSHDLMLLRLTQPAEITDAVQVVELPTQEPEVGSTCLASGWGSIEPENFS FPDDLQCVDLEILPNDECAKAHTQKVTEFMLCAGHLEGGKDTCVGDSGGPLTCD
GVLQGVTSWGYIPCGSPMKPAVFVKVLSYVKWIEDTIAENS (SEQ ID NO. 3)
A further embodiment of the present invention includes cotton top tamarin tissue kallikrein (kidney/pancreas/salivary gland kallikrein) (SEQ ID NO, 4) which has 82% sequence identity to human KLKl (SEQ ID NO, 1):
Q9N1Q1_SAGOE
MWFL VLCLALSLGGTGA VPPIQSRIVGGWDCKQHSQPWQAALYHYSTFQCGGV
LVHPQWVLTAAHCISDHYQLWLGRHDLFENEDTAQFVFVSKSFPHPDFNMSLLK NHTRLPGEDYSHDLMLLQLKQPVQITDA VK VVELPTEGIEVGSTCLASGWGSIKP EKFSFPDILQCVDLKILPNDECDKAHAQKVTEFMLCAGPLKDGQDTCVGDSGGP
LTCDGVLQGIISWGYIPCGSPNKPSVFVR VLSYVKWIKDTIADNS (SEQ ID NO, 4)
A further embodiment of the present invention includes dog tissue kallikrein (kidney/pancreas/salivary gland kallikrein) (SEQ ID NO. 5) which has 74% sequence identity to human KLK 1 (SEQ ID NO, 1 ): Q29474_CANFA
NfWFLVLCLALSLAGTGAAPPVQSRIIGGWDCTKNSQPWQAALYHYSKFQCGGV L VHPEW WTAAHC1NDNYQLWLGRYNLFEHEDT AQFVQVRESFPHPEFNLSLLK
NHTRLPEEDYSHDIMLLRLAEPAQITDAVRVLDLPTQEPQVGSTCYASGWGSIEP
DKFIYPDDLQCVDLELLSNDICANAIISQKVTEFMLCAGIILEGGKDTCVGDSGGP LICDGVLQGITSWGHVPCGSPNMPA VYTK VISHLEWIKETMTANP (SEQ ID NO.
5)
A further embodiment of the present invention includes sheep tissue kallikrein-1 (SEQ ID
NO, 6) which has 72% sequence identity to human KLKi (SEQ ID NO, 1):
A5A2L9_SHEEP
MWFPVLCLALSLAGTGA VPPVQSRIVGGQECEKHSQPWQVAIYHFSTFQCGGVL
VAPQWVLTAAHCKSENYQVWLGRHNLFEDEDTAQFAGVSEDFPNPGFNLSLLE
NΉTRQPGEDYSHDLMLLRLQEPVQLTQDVQVLGLPTKEPQLGTTCYASGWGSV
KPDEFSYPDDLQCVDLTLLPNEKCATAHPQEVTDCMLCAGHLEGGKDTCVGDS GGPLICEGMLQGITSWGHIPCGTPNKPSVYTKVIVYLDWINKTMTDNP (SEQ ID NO. 6)
A further embodiment of the present invention includes rabbit tissue kallikrein-1 (SEQ ID NO 7) which has 73% sequence identity to human KLKl (SEQ ID NO I)-
A5A2MO_RABΓΓ
MWLPVLCLALSLGGTGAAPPLQSRIIGGWVCGKNSQPWQAALYHYSNFQCGGV
LVHPQWVLTAAHCFSDNYQLWLGRHNLFEDEAEAQFIQVSGSFPHPRFNLSLLE NQTRGPGEDYSHDLMLLKLARPVQLTNAVRVLELPTQEPQVGTSCLASGWGSIT
PIKFTYPDELQCVDLSILANSECDKAHAQMVTECMLCAGHLEGGRDTCVGDSGG PL VCNNELQGITSWGHVPCGSPNKPA VFTKVLSYVEWIRNTIANNP (SEQ ID NO,
A further embodiment of the present invention includes bovine glandular kallikrein-1 precursor (SEQ ID NO. 8) which has 72% sequence identity to human KLKl (SEQ ID NO, 1):
Q6H320_BOVIN
MWFPVLCLALSLAGTGA VFPIQSRIVGGQECEKHSQPWQVAIYHFSTFQCGG VL
VAPQWVLTAAHCKSDNYQVWLGRHNLFEDEDTAQFAGVSEDFPNPGFNLSLLE
NHTRHPGEDYSHDLMLLRLQEPVQLTQNVQVLGLPTKEPQLGTTCYASGWGSV KPDEFSYPDDLQCVDLTLLPNEKCATAHPQEVTEWMLCAGHLEGGKDTCVGDS GGPLICEGMLQGITSWGHIPCGTPNKPSVYTKVILYLDWINKTMTDNP (SEQ ID NO, 8) A further embodiment of the present invention includes horse glandular kalHkrein-i precursor (KLKEl) (SEQ ID NO. 9) which has 70% sequence identity to human KLKl
(SEQ lD NO, 1):
Q6H322_HORSE NfWLPVLCLALSLVGTGAAPPIQSRIIGGWECKNHSKPWQA A VYHYSSFQCGGVL VDPQWVLTAAHCKGDYYQIWLGRHNLFEDEDTAQFFL VAKSFPHPDFNMSLLE NΗΗRLPGEDYSHDLMLLQVEQPDQITVA VQVLALPTQEP VLGSTCYASGWGSIE PDKFTYPDELRCVDLTLLSNDVCDNAHSQNVTEYMLCAGHLEGGKDTCVGDSG GPLICDGVFQGVTSWGHIPCGRPNKPAVYTKLiPHVQWlQDTIAANP (SEQ ID NO, 9)
A preferred embodiment of the present invention includes pig glandular kallikrein-i precursor (SEQ ID NO, 10) which has 67% sequence identity to human KLKl (SEQ ID NO, 1):
NPJ)Q 100191 1 GI.-50054435 Sm scrofa
1-17 signal peptide 18-24 propeptide 25-263 mature peptide
>gi|50054435|ref|NP_00100191 1.1 1 kallikrein 1 [Sus scrofa]
MW SLVMRLALSLAGTGAAPPIQSRIIGGRECEKDSHPWQVA IYHYS SFQCGGVL
VDPKWVLTAAHCKNDNYQVWLGRHNLFENEVTAQFFGVTADFPHPGFNLSLLK NHTKADGKDYSHDLMLLRLQSPAKITDAVKVLELPTQEPELGSTCQASGWGSIE PGPDDFEFPDEIQCVELTLLQNTFCADAHPDKVTESMLCAGYLPGGKDTCMGDS GGPLICNGJVIWQGITSWGHTPCGSANKPSIYTKLIFYLDWINDTITENP (SEQ ID NO, 10)
The term "active fragment" refers to smaller portions of the KLKl polypeptide that retains the activity of the full-length KLKl polypeptide, for example, the KLKl without the signal peptide region, the KLKl without the signal peptide and without the propeptide regions, and fragments of the KLKl protein found to have serine protease activity capable of cleaving low-molecular weight kininogen into kallidin.
A "variant" or "mutant" of a starting or reference polypeptide is a polypeptide that 1) has an amino acid sequence different from that of the starting or reference polypeptide and 2) was derived from the starting or reference polypeptide through either natural or artificial (manmade) mutagenesis. Such variants include, for example, deletions from, and/or insertions into and/or substitutions of, residues within the amino acid sequence of the polypeptide of interest, A variant amino acid, in this context, refers to an amino acid different from the amino acid at the corresponding position in a starting or reference polypeptide sequence {such as that of a source antibody or antigen binding fragment). Any combination of deletion, insertion, and substitution may be made to arrive at the final variant or mutant construct, provided that the final construct possesses the desired functional characteristics. The amino acid changes also may alter post-translational processes of the polypeptide, such as changing the number or position of glycosylation sites. Methods for generating amino acid sequence variants of polypeptides are described in U.S. Patent No, 5,534,615, expressly incorporated herein by reference. Variants or mutants of a reference polypeptide may, for example, have 95, 90, 85, 82, 80, 75, 74, 72, or 60% identity with said reference polypeptide and may have more, less, or identical activity to the reference polypeptide. Variants may also include sequences added to the reference polypeptide to facilitate purification, to improve metabolic half-life or to make the polypeptide easier to identify, for example, a His tag or a pegylation sequence.
A "wild type" or "reference" sequence or the sequence of a "wild type" or "reference" protein/poly peptide maybe the reference sequence from which variant polypeptides are derived through the introduction of mutations. In general, the "wild type" sequence for a given protein is the sequence that is most common in nature. Similarly, a "wild type" gene sequence is the sequence for that gene which is most commonly found in nature. Mutations may be introduced into a "wild type" gene (and thus the protein it encodes) either through natural processes or through man induced means. The products of such processes are "variant" or "mutant" forms of the original "wild type" protein or gene.
"Percent (%) amino acid sequence identity" with respect to the polypeptides identified herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. The ALIGN-2 program is publicly available through Genentech, Inc., South San Francisco, California.
For purposes herein, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows:
100 times the fraction X/Y, where X is the number of amino acid residues scored as identical matches by the sequence alignment program in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A, "Percent (%) nucleic acid sequence identity" is defined as the percentage of nucleotides in a candidate sequence that are identical with the nucleotides in a reference poly pepti de-encoding nucleic acid sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent nucleic acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared can be determined by known methods. For purposes herein, the % nucleic acid sequence identity of a given nucleic acid sequence C to, with, or against a given nucleic acid sequence D (which can alternatively be phrased as a given nucleic acid sequence C that has or comprises a certain % nucleic acid sequence identity to, with, or against a given nucleic acid sequence D) is calculated as follows:
100 times the fraction W/Z, where W is the number of nucleotides scored as identical matches by the sequence alignment program in that program's alignment of C and D, and where Z is the total number of nucleotides in D. It will he appreciated that where the length of nucleic acid sequence C is not equal to the length of nucleic acid sequence D, the % nucleic acid sequence identity of C to D will not equal the % nucleic acid sequence identity of D to C. The term "amino acid" is used in its broadest sense and is meant to include the naturally occurring L α-amino acids or residues. The commonly used one and three letter abbreviations for naturally occurring amino acids are used herein (Lehninger, A. L., Biochemistry, 2d ed., pp. 71-92, (1975), Worth Publishers, New York). The term also includes all D-amino acids as well as chemically modified amino acids such as amino acid analogs, naturally occurring amino acids that are not usually incorporated into proteins such as Norleucine, and chemically synthesized compounds having properties known in the art to be characteristic of an amino acid. For example, analogs or mimetics of phenylalanine or proline, which allow the same conformational restriction of the peptide compounds as natural Plie or Pro are included within the definition of amino acid. Such analogs and mimetics are referred to herein as
"functional equivalents" of an amino acid. Other examples of amino acids are listed by Roberts and Vellaccio, In: The Peptides: Analysis, Synthesis, Biology, Gross and Meiehofer, Eds., Vol. 5 p 341, Academic Press, Inc, N. Y. 1983, which is incorporated herein by reference. The term "protein" has an amino acid sequence that is longer than a peptide. A
"peptide" contains 2 to about 50 amino acid residues. The term "polypeptide" includes proteins and peptides. Examples of proteins include, but are not limited to, antibodies, enzymes, lectins and receptors; lipoproteins and lipopolypeptides; and glycoproteins and gl y copol y pepti des. A "fusion protein" and a "fusion polypeptide" refer to a polypeptide having two portions covalently linked together, where each of the portions is a polypeptide having a different property. The property may be a biological property, such as activity in vitro or in vivo. The property may also be a simple chemical or physical property, such as binding to a target antigen, catalysis of a reaction, etc. The two portions may be linked directly by a single peptide bond or through a peptide linker containing one or more amino acid residues. Generally, the two portions and the linker will be in reading frame with each other. Preferably, the two portions of the polypeptide are obtained from heterologous or different polypeptides.
The term "therapeutically effective amount" refers to an amount of a composition of this invention effective to "alleviate" or "treat" a disease or disorder in a subject or mammal. Generally, alleviation or treatment of a disease or disorder involves the lessening of one or more symptoms or medical problems associated with the disease or disorder. In some embodiments, it is an amount that can increase β -cell mass as compared to the diseased state.
The terms "treatment" and "treating" refer to inhibiting, alleviating, and healing disease, conditions or symptoms thereof, "Treating" or "treatment" refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) the targeted pathologic condition or disorder. Treatment can be carried out by administering a therapeutically effective amount of at least one compound of the invention. Parameters for assessing successful treatment and improvement in the disease are readily measurable by routine procedures familiar to a physician.
The term "β-cell regeneration", or 'increase in β-cell mass" refers to enhanced β- cell mass in the pancreas as determined by a physician skilled in the art through methods of imaging methods and metabolic assessment. Imaging methods include, but are not limited to, positron emission spectroscopy,
MRI and/or single photon emission computed tomography. Metabolic assessment includes, but is not limited to, the oral glucose tolerance test, the intravenous glucose tolerance test, the intravenous arginine stimulation test and glucose potentiation of argi nine-induced insulin secretion. Known mathematic models for assessing β-cell function include, but are not limited to the homeostatic model assessment (HOMA) and continuous infusion of glucose with model assessment (CIGMA). HOMA uses a structural model of glucose-insulin interaction and assesses β-cell function and insulin resistance from basal glucose and insulin or C-peptide concentrations in a patient (Wallace, 2004), ClGMA evaluates the near steady state glucose/insulin concentration after approximately 1-2 hours, continuous glucose infusion that causes plasma glucose levels similar to postpradial levels (Hermans, 1999)
Methods of Modulating Pancreatic Islet β-Cell Mass
The present invention provides methods for treating pancreatic islet β-cell dysfunction and diseases and conditions associated therewith. One embodiment includes a method of treatment of pancreatic islet cell dysfunction and treating diseases and conditions associated therewith by the modulation β-cell mass in a mammal by administering to the mammal a therapeutically effective amount of KLKl . Pharmaceutical compositions may be administered orally, Oral administration includes enteral administration of solution, tablets, sustained release capsules, enteric coated capsules, and syrups.
An "effective amount" or a "therapeutically effective amount" refers to a nontoxic but sufficient amount of drug or agent to provide a desired effect, In a combination therapy, an "effective amount" of one component of the combination is an amount of that compound that is effective to provide a desired effect when used in combination with the other components of the combination. An amount that is "effective" will vary from subject to subject, depending on the age and general condition of an individual, a particular active agent or agents, and the like. An appropriate "effective" amount in any individual case may be determined using routine experimentation. A therapeutically effective amount of a compound of the invention for treating the above-identified diseases or symptoms thereof can be administered prior to, concurrently with, or after the onset of the disease or symptom, A compound of the invention can be administered concurrently with the onset of the disease or symptom, "Concurrent administration" and "concurrently administering" as used herein includes administering a polypeptide of the invention and another therapeutic agent in admixture, such as, for example, in a pharmaceutical composition or in solution, or separately, such as, for example, separate pharmaceutical compositions or solutions administered consecutively, simultaneously, or at different times, but not so distant in time such that the compound of the invention and the other therapeutic agent cannot interact and a lower dosage amount of the active ingredient cannot be administered. Another aspect of the present invention includes a method as herein described further comprising concurrently using an additional therapeutic method useful in the treatment of pancreatic islet cell dysfunction and treating disease and symptoms associated therewith. An additional therapeutic method includes, but is not limited to, an stem cell transplant and pancreatic organ transplant. "Treatment" and "treating" refer to preventing, inhibiting, and/or alleviating disease or symptoms associated with pancreatic islet cell dysfunction as well as healing disease or symptoms associated with pancreatic islet cell dysfunction affecting mammalian organs and tissues. A composition of the present invention can be administered in a therapeutically effective amount to a patient before, during, and after any above-mentioned condition arises.
The invention will be described with reference to various specific and preferred embodiments and techniques. However, it should be understood that many variations and modifications may be made while remaining within the spirit and scope of the invention,
EXAMPLES
The methods employed herein are similar to those seen in the works of Ellingsgaard (Ellingsgaard et a/, PNAS, 2008, 105(35): 13162-7).
Example 1: Modulation of Pancreatic Islet Cell Mass
20 Male C57BL/6J mice aged 8 weeks were fed a hypercaloric diet consisting of 58% calories from fat for 8 weeks. Water was provided ad libitum. The mice are genetically predisposed to developing type 2 diabetes and are often used for diabetes and obesity studies. 10 mice in the treatment group were injected intramuscularly ( 2ml/kg) with isolated pig KLK 1 having a sequence of SEQ ID NO, 10 at a dosage of 0,375 mg/ml in PBS and daily the control group received a placebo injection of PBS. For the final 14 days the mice were exposed to 1 mg/ml of bromodeoxyuridine (BrdU) in the drinking water, which is used to detect the proliferation of the beta cells.
Animals were sacrificed. The pancreas of each animal was harvested and fixed in paraformaldehyde. Histological analysis was done. The 20 pancreas samples were stained with anti-BrdU, to detect cell proliferation and anti-insulin to detect beta cells.
Results
Data was interpreted manually by a technician. Staining was graded as either weak (1); moderate (2); or strong (3), The percent of the beta cells stained was also recorded. Cells treated with KLKl showed higher beta cell intensity on average (40% more intensity) and a higher percentage of BrdU in the beta cells (45% more proliferation) as compared to the control cells. Therefore KLKl is able to trigger β-stem cell proliferation (Table 1 , and Table 2.)
Table 1: Effect of PBS on beta cell intensit and BrdU stainin
Figure imgf000017_0001
Table 2: Effect of KLKI on beta cell intensity and BrdU staining
Figure imgf000017_0002
Figure imgf000018_0001
Example 2: Treatment of Streptozotocin treated rats with KLKl
MaIeWi star rats 7-9 weeks old (225-275 grams) were used. The animals were fed a standard Purina chow diet. The duration of the study was 28 days but 3 weeks of treatment. The rats were divided into 5 treatment groups: (a) no STZ (n=8), (b) PBS vehicle (n=8), (c) KLKl 0.2U (n=8), (d) KLKl 1 U (n=8) and (e) KLKl 5LJ (n=8). The animals were injected once with 50 mg/kg of streptozotocin (STZ) to cause beta cell death as STZ is a beta cell specific toxin, KLKl treated animals had BID IP injections on days 7 to 28. All animals had daily IP injections of bromodeoxyuridine (BrdU) (50 mg/kg) on days 7 to 15, BrdU is used for the detection of beta cells.
The animals were sacrificed. Formalin-fixed pancreas samples were embedded in paraffin and sectioned at approximately 5 microns. One serial section was stained with hematoxylin and eosin (H&E) and another serial section underwent an immunohistochemical (IHC) protocol. This included an ant-BRDU antibody conjugated to DAB as a chromagen (brown) and an anti -insulin antibody conjugated to fast red as a chromagen All slides were examined by a board -certified veterinary pathologist The following parameters were assessed:
Pancreatic Beta Cell Area:
For each IHC sample 2 typical 4x objective fields were photographed at a fixed pixel density that contained all or mostly all pancreas with representative areas of islets. Using Nikon Elements 3,0 software these images were thresholded for insulin-positive tissue (red tissue) and the number of insulin-positive pixels was recorded. The image was then thresh olded for all pancreatic tissue present (excluding white space and all non -pancreatic tissue such as small intestine or lymph node) and the number of pixels here recorded.
The area of insulin-positive tissue was then reported as the Area of insulin positive tissue / Area of all pancreatic tissue in the image. The 2 such data points for each sample were summed and this data was presented.
Islet Number:
In these same two 4x objective fields per sample the number of islets was counted and recorded.
Pancreatic Beta Cell Replication:
Brown BrdU-positive nuclei that were within insulin-positive cells of islets were counted, The number of such cells in 5 evenly- sized large islets was counted and recorded.
Number of Insulin-Positive Duct Cells:
Five 2Ox objective fields with large pancreatic ducts were assessed. Any insulin-positive duct cells in these 5 fields were counted and recorded.
Results:
Figure 1 shows Pancreatic Beta Cell Area for the treatment groups. Pancreatic Beta Cell area was reduced in the STZ + PBS group compared to the No STZ group indicating that streptozotocin has successfully induced islet atrophy, as expected.
All three dose groups of Compound 1 (KLKl) notably increase pancreatic beta cell area as compared to the STZ + PBS group. Figure 2 shows islet number data for the treatment groups. Islet number was notably reduced in the STZ + PBS group compared to the No STZ group indicating that streptozotocin successfully induced islet atrophy, as expected,
All three dose groups of Compound 1 (KLKl) notably increase islet number compared to the STZ + PBS group.
Figure 3 shows data regarding beta cell replication in islets. Beta cell replication is notably reduced in the STZ + PBS group compared to the No STZ group indicating that streptozotocin has successfully hindered beta cell replication in islets, as expected.
All three dose groups of Compound 1 notably increase this parameter compared to the STZ + PBS group, and the 0.2U dose does so in a statistically significant manner (as shown in Figure 3, ** = p<G,01).
Figure 4 shows the number of insulin-positive duct cells in the pancreas for all treatment groups. The number of insulin-positive duct cells was slightly reduced in the STZ + PBS group compared to the No STZ group,
All three dose groups of Compound 1 (KLKl ) notably increase the number of insulin-positive duct cells in the pancreas, compared to the STZ + PBS group and the No STZ group.
Figure 5 shows representative images of pancreatic islets, for the five treatment groups (No STZ, STZ + PBS, STZ + Compound 1 (KLKl) at 0.2 units, STZ + Compound 1 (KLKl) at 1,0 units, and STZ+ Compound 1 (KLKl) at 5,0 Units, Pancreatic islets were stained bright red via insulin IHC. Images were taken at 4Ox magnification.

Claims

1. A method of treating pancreatic islet β-cell dysfunction comprising administering a therapeutically effective amount of a purified or isolated KLKl, or a variant or active fragment thereof,
2. The method of claim 1 wherein the purified or isolated KLK 1 , or variant or active fragment thereof, is a purified and isolated natural KLKl,
3. The method of claim 1 wherein the purified or isolated KLK 1 , or variant or active fragment thereof, is recombinantly produced,
4. The method of any one of claims 1-3 wherein the purified or isolated KLK 1 comprises the amino acid sequence of SEQ ID NO. 1.
5. The method of any one of claims 1-3 wherein the purified or isolated KLK 1 comprises an amino acid sequence selected from the group consisting of SEQ ID NO, 2, SEQ ID NO, 3, SEQ ID NO, 4, SEQ ID NO, 5, SEQ ID NO, 6, SEQ ID
NO. 7, SEQ ID NO. 8, SEQ ID NO. 9, and SEQ ID NO. 10.
6. The method of any one of claims 1-3 wherein the purified or isolated KLK 1 comprises a polypeptide having 90% sequence identity to SEQ ID NO, 1 and serine protease activity capable of cleaving low molecular weight kininogen into kallidin,
7. The method of any one of claims 1-3 wherein the purified or isolated KLK 1 comprises a polypeptide having 82% sequence identity to SEQ ID NO. 1 and serine protease activity capable of cleaving low molecular weight kininogen into kallidin.
8, The method of any one of claims 1-3 wherein the purified or isolated KLK l comprises a polypeptide having 74% sequence identity to SEQ ID NO. 1 and serine protease activity capable of cleaving low molecular weight kininogen into kallidin,
9. The method of any one of claims 1 -3 wherein the purified or isolated KLK 1 comprises a polypeptide having 72% sequence identity to SEQ ID NO, 1 and serine protease activity capable of cleaving low molecular weight kininogen into kallidin.
10. The method of any one of claims 1-3 wherein the purified or isolated KLK l comprises a polypeptide having 60% sequence identity to SEQ ID NO. 1 and serine protease activity capable of cleaving low molecular weight kininogen into kallidin,
1 1. The method of claim 3 wherein the purified or isolated KLKl or variant or active fragment thereof comprises a pegylation sequence,
12. The method of any one of claims 1-1 1, wherein the pancreatic islet β-cell dysfunction comprises an abnormal β-cell mass,
13. A method of any one of claims 1-12, wherein the purified or isolated KLKl, or a variant or active fragment thereof, is administered orally.
14. A method of any one of claims 1-13, wherein the purified or isolated KLKl, or a variant or active fragment thereof, is administered concurrently with a second therapeutic method useful in treating pancreatic islet cell dysfunction,
15. A method of claim 14, wherein the second therapeutic method is selected from the group consisting of a stem cell transplant, and a pancreatic organ transplant.
16. The method of any one of claims 1-15 whereby the treatment modulates β-cell mass,
17, The method of claim 16 wherein the modulation of β-cell mass is selected from the group consisting of an increase in β-cell mass, β-cell regeneration, a restoring of normal β-cell function by increasing the number of functional β-cell s, an increase in β-cell proliferation, and restoring or improving function in impaired β- cells.
18, Use of a purified or isolated KLK 1, or a variant or active fragment thereof for the treatment of pancreatic islet β-cell dysfunction.
19, Use of a purified or isolated KLKl, or a variant or active fragment thereof in the preparation of a medicament for the treatment of pancreatic islet β-cell dysfunction.
20, Use of a purified or isolated KLKl, or a variant or active fragment thereof, in the preparation of a medicament for the treatment of pancreatic islet β-cell dysfunction,
21 , Use of claim 20 wherein the purified or isolated KLKl, or variant or active fragment thereof, is a purified and isolated natural KLKl,
22, Use of claim 20 wherein the purified or isolated KLKl, or variant or active fragment thereof, is recombinantly produced.
23, Use of claim 20, wherein the purified or isolated KLKl comprises the amino acid sequence of SEQ ID NO. 1.
24, Use of claim 20, wherein wherein the purified or isolated KLKl comprises an amino acid sequence selected from the group consisting of SEQ ID NO, 2, SEQ ID NO. 3, SEQ ID NO. A, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 7, SEQ ID NO. 8, SEQ ID NO. 9, and SEQ ID NO. 10.
25, Use of claim 20, wherein the purified or isolated KLKl comprises a polypeptide having 90% sequence identity to SEQ ID NO. 1 and serine protease activity capable of cleaving low molecular weight kininogen into kallidin.
26. Use of claim 20, wherein the purified or isolated KLKl comprises a polypeptide having 82% sequence identity to SEQ ID NO. 1 and serine protease activity capable of cleaving low molecular weight kininogen into kallidin,
27, Use of claim 20, wherein the purified or isolated KLKl comprises a polypeptide having 74% sequence identity to SEQ ID NO. 1 and serine protease activity capable of cleaving low molecular weight kininogen into kallidin.
28. Use of claim 20, wherein the purified or isolated KLKl comprises a polypeptide having 72% sequence identity to SEQ ID NO. 1 and serine protease activity capable of cleaving low molecular weight kininogen into kallidin,
29. Use of claim 20 wherein the purified or isolated KLKl comprises a polypeptide having 60% sequence identity to SEQ ID NO. 1 and serine protease activity capable of cleaving low molecular weight kininogen into kallidin,
30. Use of claim 20 wherein the purified or isolated KLKl or variant or active fragment thereof comprises a pegylation sequence. , Use of claim 20 wherein the pancreatic islet β-cell dysfunction comprises an abnormal β-cell mass. , Use of claim 20 wherein the purified or isolated KLKl, or a variant or active fragment thereof, is administered orally.
PCT/CA2010/000413 2009-03-25 2010-03-25 TISSUE KALLIKREIN FOR THE TREATMENT OF PANCREATIC β-CELL DYSFUNCTION WO2010108262A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
NZ595364A NZ595364A (en) 2009-03-25 2010-03-25 TISSUE KALLIKREIN FOR THE TREATMENT OF PANCREATIC beta-CELL DYSFUNCTION
EP10755352A EP2411042A4 (en) 2009-03-25 2010-03-25 Tissue kallikrein for the treatment of pancreatic -cell dysfunction
AU2010228068A AU2010228068A1 (en) 2009-03-25 2010-03-25 Tissue kallikrein for the treatment of pancreatic beta-cell dysfunction
JP2012501089A JP2012521366A (en) 2009-03-25 2010-03-25 Tissue kallikrein for the treatment of pancreatic beta cell dysfunction
CA2756801A CA2756801A1 (en) 2009-03-25 2010-03-25 Tissue kallikrein for the treatment of pancreatic .beta.-cell dysfunction
CN2010800181825A CN102438648A (en) 2009-03-25 2010-03-25 Tissue kallikrein for the treatment of pancreatic beta-cell dysfunction
US13/241,882 US20120070425A1 (en) 2009-03-25 2011-09-23 TISSUE KALLIKREIN FOR THE TREATMENT OF PANCREATIC Beta-CELL DYSFUNCTION AND FOR Beta-CELL PROLIFERATION

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US16317309P 2009-03-25 2009-03-25
US61/163,173 2009-03-25

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/241,882 Continuation-In-Part US20120070425A1 (en) 2009-03-25 2011-09-23 TISSUE KALLIKREIN FOR THE TREATMENT OF PANCREATIC Beta-CELL DYSFUNCTION AND FOR Beta-CELL PROLIFERATION

Publications (1)

Publication Number Publication Date
WO2010108262A1 true WO2010108262A1 (en) 2010-09-30

Family

ID=42780097

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2010/000413 WO2010108262A1 (en) 2009-03-25 2010-03-25 TISSUE KALLIKREIN FOR THE TREATMENT OF PANCREATIC β-CELL DYSFUNCTION

Country Status (8)

Country Link
US (1) US20120070425A1 (en)
EP (1) EP2411042A4 (en)
JP (1) JP2012521366A (en)
CN (1) CN102438648A (en)
AU (1) AU2010228068A1 (en)
CA (1) CA2756801A1 (en)
NZ (1) NZ595364A (en)
WO (1) WO2010108262A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8501695B2 (en) 2007-07-20 2013-08-06 Diamedica, Inc. Tissue kallikrein for the treatment of diseases associated with amyloid protein
CN103889435A (en) * 2011-06-17 2014-06-25 约翰斯·霍普金斯大学 Methods for increasing insulin sensitivity and treating diabetes
WO2015100768A1 (en) * 2013-12-30 2015-07-09 江苏众红生物工程创药研究院有限公司 Pegylated tissue kallikrein, and preparation method therefor and uses thereof
US9364521B2 (en) 2012-06-04 2016-06-14 Diamedica Inc. Human tissue kallikrein 1 glycosylation isoforms
US9616015B2 (en) 2012-05-25 2017-04-11 Diamedica Inc. Formulations of human tissue kallikrein-1 for parenteral delivery and related methods
US11857608B2 (en) 2017-03-09 2024-01-02 Diamedica Inc. Dosage forms of tissue kallikrein 1

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112481268B (en) * 2021-01-25 2024-01-30 河南大学 Cotton promoter P GhPGF And recombinant vector and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5534615A (en) 1994-04-25 1996-07-09 Genentech, Inc. Cardiac hypertrophy factor and uses therefor
US20040253226A1 (en) * 1998-05-22 2004-12-16 Holaday John W. Compositions and methods for inhibiting endothelial cell proliferation and regulating angiogenesis using serine proteases

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1228447C (en) * 2001-02-20 2005-11-23 深圳市人民医院 Recombinant expression vector expressing human pancreatic tissue kallikrein gene and prepn of human pancreatic tissue kallikrein
CN100338212C (en) * 2003-01-29 2007-09-19 中国科学院大连化学物理研究所 Microencapsulated cell of human tissue kallikrein, microencapsulating method and application thereof
CN101094869A (en) * 2004-08-03 2007-12-26 戴埃克斯有限公司 Hk1-binding proteins
CA2575791A1 (en) * 2004-08-03 2006-02-16 Dyax Corp. Hk1-binding proteins
EP2051732A4 (en) * 2006-07-26 2009-10-21 Diamedica Inc Methods of diagnosis and treatment for metabolic disorders
CA2658523C (en) * 2006-07-31 2012-06-12 Activesite Pharmaceuticals, Inc. Inhibitors of plasma kallikrein
CN101134953B (en) * 2007-07-02 2011-02-09 广东天普生化医药股份有限公司 Recombinant human pancreas kininogenase
CN101255438B (en) * 2008-04-11 2012-01-25 深圳大学 Construction method of transgenic chlamydomonas reinhardtii for expressing human tissue kallikrein

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5534615A (en) 1994-04-25 1996-07-09 Genentech, Inc. Cardiac hypertrophy factor and uses therefor
US20040253226A1 (en) * 1998-05-22 2004-12-16 Holaday John W. Compositions and methods for inhibiting endothelial cell proliferation and regulating angiogenesis using serine proteases

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
CLEMENTS, J. ET AL.: "The expanded human kallikrein (KLK) gene family: Genomic organization, tissue-specific expression and potential functions.", BIOLOGICAL CHEMISTRY., vol. 382, no. 1, January 2001 (2001-01-01), pages 5 - 14 *
DONG, Y. ET AL.: "Tissue-specific promoter utilisation of the kallikrein-related peptidase genes, KLK5 and KLK7, and cellular localisation of the encoded proteins suggest roles in exocrine pancreatic function.", BIOLOGICAL CHEMISTRY., vol. 389, no. 2, February 2008 (2008-02-01), pages 99 - 109 *
ELLINGSGAARD ET AL., PNAS, vol. 105, no. 35, 2008, pages 13162 - 7
EMAMI, N. ET AL.: "Utility of Kallikrein-related peptidases (KLKs) as cancer biomarkers.", CLINICAL CHEMISTRY, vol. 54, no. 10, October 2008 (2008-10-01), pages 1600 - 1607 *
LEHNINGER A.L.: "Biochemistry", 1975, WORTH PUBLISHERS, pages: 71 - 92
MEIER, DIABETOLOGIA, vol. 51, 2008, pages 703 - 13
MUELLER, PHYS THER, vol. 88, no. 11, 2008, pages 1250 - 3
ROBERTS, VELLACCIO: "The Peptides: Analysis, Synthesis, Biology", vol. 5, 1983, ACADEMIC PRESS, INC, pages: 341
See also references of EP2411042A4 *
SHAW, J. ET AL.: "Regulation of human tissue kallikrein-related peptidase expression by steroid hormones in 32 cell lines.", BIOLOGICAL CHEMISTRY., vol. 389, no. 11, November 2008 (2008-11-01), pages 1409 - 1419 *
YOUSEF ET AL., ENDOCRINE REV., vol. 22, 2001, pages 184 - 204
YOUSEF, G. ET AL.: "Genomic organization of the human kallikrein gene family on chromosome 19q13.3-q13.4.", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS., vol. 276, no. 1, September 2000 (2000-09-01), pages 125 - 133 *
YOUSEF, G. ET AL.: "In-silico analysis of kallikrein gene expression in pancreatic and colon cancers.", ANTICANCER RESEARCH., vol. 24, no. 1, January 2004 (2004-01-01), pages 43 - 51 *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8501695B2 (en) 2007-07-20 2013-08-06 Diamedica, Inc. Tissue kallikrein for the treatment of diseases associated with amyloid protein
CN103889435A (en) * 2011-06-17 2014-06-25 约翰斯·霍普金斯大学 Methods for increasing insulin sensitivity and treating diabetes
JP2014520119A (en) * 2011-06-17 2014-08-21 ザ・ジョンズ・ホプキンス・ユニバーシティー Methods for increasing insulin sensitivity and methods for treating diabetes
EP2720703A4 (en) * 2011-06-17 2015-07-22 Univ Johns Hopkins Methods for increasing insulin sensitivity and treating diabetes
US9616015B2 (en) 2012-05-25 2017-04-11 Diamedica Inc. Formulations of human tissue kallikrein-1 for parenteral delivery and related methods
US9364521B2 (en) 2012-06-04 2016-06-14 Diamedica Inc. Human tissue kallikrein 1 glycosylation isoforms
US9839678B2 (en) 2012-06-04 2017-12-12 Diamedica Inc. Human tissue kallikrein 1 glycosylation isoforms
WO2015100768A1 (en) * 2013-12-30 2015-07-09 江苏众红生物工程创药研究院有限公司 Pegylated tissue kallikrein, and preparation method therefor and uses thereof
US10052368B2 (en) 2013-12-30 2018-08-21 Zonhon Biopharma Institute Inc. Pegylated tissue kallikrein, and preparation method therefor and uses thereof
US11857608B2 (en) 2017-03-09 2024-01-02 Diamedica Inc. Dosage forms of tissue kallikrein 1

Also Published As

Publication number Publication date
CA2756801A1 (en) 2010-09-30
US20120070425A1 (en) 2012-03-22
NZ595364A (en) 2013-09-27
JP2012521366A (en) 2012-09-13
EP2411042A4 (en) 2012-12-12
AU2010228068A1 (en) 2011-10-20
EP2411042A1 (en) 2012-02-01
CN102438648A (en) 2012-05-02

Similar Documents

Publication Publication Date Title
WO2010108262A1 (en) TISSUE KALLIKREIN FOR THE TREATMENT OF PANCREATIC β-CELL DYSFUNCTION
CN103402536A (en) Methods for treating metabolic disorders and obesity with GIP and GLP-1 receptor-active glucagon-based peptides
CN101914150B (en) Polypeptide and use thereof in medicament preparation
JP2014534265A (en) Therapeutic drugs containing insulin amino acid sequence
Suzuki et al. Recent developments in therapeutic peptides for the glucagon-like peptide 1 and 2 receptors
IL99699A (en) Pharmaceutical oral, enteral or by-inhalation dosage form for suppressing an autoimmune response associated with type i diabetes
TWI714862B (en) A method and drug for regulating GLP-1/GLP-1R
CN105188736A (en) Insulin dosing regimen
KR20210024583A (en) How to use GIP/GLP1 coagonists for therapy
KR20160075794A (en) Calcitonin mimetics for treating diseases and disorders
US20200338158A1 (en) Therapeutic Drug for Non-Alcoholic Fatty Liver Disease
CN109125715A (en) A kind of method and drug of regulation GLP-1/GLP-1R
Boj-Carceller Proton pump inhibitors: impact on glucose metabolism
CN105579052A (en) Pharmaceutical composition
EP0835129B1 (en) Prevention of a disease having the characteristics of diabetes
CN110167582A (en) A method of preventing and treating Drug injury of kidney
US7795385B2 (en) Use of bombesin/gastrin-releasing peptide antagonists for the treatment of inflammatory conditions, acute lung injury and bipolar disorder
CN102292100A (en) Adiponectin for treating pulmonary disease
US20230414706A1 (en) Compositions and methods for treating metabolic diseases
Shaikh et al. Advanced approaches in insulin delivery
US8895505B2 (en) Method of treatment of type 2 diabetes
CN112826924B (en) Use of an intestine-targeted gastrin-silica complex
CN100508957C (en) Improved oral delivery of peptides
JPH0680584A (en) Medicine containing glycentin as active ingredient
Do SunGil et al. Silk fibroin hydrolysate exerts an anti-diabetic effect by increasing pancreatic β cell mass in C57BL/KsJ-db/db mice.

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201080018182.5

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10755352

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2012501089

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2756801

Country of ref document: CA

Ref document number: 595364

Country of ref document: NZ

Ref document number: 2010228068

Country of ref document: AU

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2010228068

Country of ref document: AU

Date of ref document: 20100325

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 4362/KOLNP/2011

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2010755352

Country of ref document: EP