INHIBITORS OF EXTRACELLULAR PROTEASES

Provided is a plant derived extract including inhibitory activity against one or more extracellular proteases which degrade human tissue matrix. Moreover, the amount of inhibitory activity in an extract can be increased by stressing the plant prior to forming an extract. These extracts are each prepared by a process and demonstrate the ability to inhibit one or more extracellular proteases which degrade human tissue matrix. Libraries of extracts can be prepared from stressed and non-stressed plants, where each of the extracts demonstrate inhibitory activity against on or more extracellular protease inhibitors. Alternatively, semi-purified and purified inhibitory compounds can be isolated from the extracts. In one aspect, these extracts with inhibitory activity can be used during protein purification to minimize degradation due to extracellular proteases.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO EARLIER FILED APPLICATIONS

This is a Continuation Application of U.S. patent application Ser. No. 11/878,978, filed Jul. 30, 2007, which was a Continuation Application of U.S. patent application Ser. No. 10/469,402, filed Aug. 29, 2003 and claims benefit of International Application No. PCT/CA02/00285 filed Mar. 4, 2002, the contents of each of which are hereby incorporated by reference in their entirety.

FIELD OF INVENTION

The invention pertains to the field of protease inhibitors, specifically inhibitors of extracellular proteases.

BACKGROUND OF THE INVENTION

The cells of tissues are generally in contact with a network of large extracellular macromolecules that occupies the spaces in a tissue between the component cells and also occupies the space between adjacent tissues. This extracellular matrix functions as a scaffolding on which the cells and tissue are supported and is involved actively in regulating interaction of the cells that contact it. The principal macromolecules of the extracellular matrix include the collagens (the most abundant proteins in the body) and glycosaminoglycans (complex polysaccharides which are usually bonded also to protein and then termed proteoglycans). The macromolecules that comprise the extracellular matrix are produced typically by the cells in contact therewith, for example, epithelial cells in contact with a basement membrane and fibroblasts embedded in connective tissue.

The glycosaminoglycan (proteoglycan) molecules form a highly hydrated matrix (a gel) in which elastic or fibrous proteins (such as collagen fibers) are embedded. The aqueous nature of the gel permits diffusion of metabolically required substances between the cells of a tissue and between tissues. Additional proteins that may be found in extracellular matrix include elastin, fibronectin and laminin.

The term “connective tissue” refers to extracellular matrix plus specialised cells such as, for example, fibroblasts, chondrocytes, osteoblasts, macrophages and mast cells found therein. The term “interstitial tissue” is best reserved for an extracellular matrix that stabilizes a tissue internally, filling the gaps between the cells thereof. There are also specialized forms of extracellular matrix (connective tissue) that have additional functional roles—cornea, cartilage and tendon, and when calcified, the bones and teeth.

A structural form of extracellular matrix is the basal lamina (basement membrane). Basal laminae are thin zones of extracellular matrix that are found under epithelium or surrounding, for example, muscle cells or the cells that electrically insulate nerve fibres. Generally speaking, basal laminae separate cell layers from underlying zones of connective tissue or serve as a boundary between two cell layers wherein a basal lamina can serve as a pathway for invading cells associated with pathologic processes, or for structural organisation associated with tissue repair (i.e. as a blueprint from which to regenerate original tissue architecture and morphology).

The regulated turnover of extracellular matrix macromolecules is critical to a variety of important biological processes. Localised degradation of matrix components is required when cells migrate through a basal lamina, as when white blood cells migrate across the vascular basal lamina into tissues in response to infection or injury, or when cancer cells migrate from their site of origin to distant organs via the bloodstream or lymphatic vessels, during metastasis. In normal tissues, the activity of extracellular proteases is tightly regulated and the breakdown/production of connective tissue is in dynamic equilibrium, such that there is a slow and continual turnover due to degradation and resynthesis in the extracellular matrix of adult animals.

In each of these cases, matrix components are degraded by extracellular proteolytic enzymes that are secreted locally by cells. These proteases belong to one of four general classes: many are metalloproteinases, which depend on bound Ca2+ or Zn2+ for activity, while the others are serine, aspartic and cysteine proteases, which have a highly reactive serine, aspartate or cysteine residue in their respective active site (Vincenti et al., (1994) Arthritis and Rheumatism, 37: 1115-1126). Together, metalloproteinases, serine, aspartate and cysteine proteases cooperate to degrade matrix proteins such as collagen, laminin, and fibronectin.

Several mechanisms operate to ensure that the degradation of matrix components is tightly controlled. First, many proteases are secreted as inactive precursors that can be activated locally. Second, the action of proteases is confined to specific areas by various secreted protease inhibitors, such as the tissue inhibitors of metalloproteases and the serine protease inhibitors known as serpins. These inhibitors are specific for particular proteases and bind tightly to the activated enzyme to block its activity. Third, many cells have receptors on their surface that bind proteases, thereby confining the enzyme to where it is needed.

Many pathogenic bacteria produce extracellular metalloproteases, of which many are zinc containing proteases that can be classified into two families, the thermolysin (neutral) proteases and the serralysin (alkaline) proteases.

A number of patents and publications report the inhibition of one or more extracellular proteases by compounds extracted from plants. For example, Sun et al., (1996) Phytotherapy Res., 10: 194-197, reports the inhibition in vitro of stromelysin (MMP-3) and collagenase by betulinic acid extracted from Doliocarpus verruculosis. Sazuka et al, (1997).

Biosci. Biotechnol. Biochem., 61: 1504-1506, reports the inhibition of gelatinases (MMP-2 and MMP-9) and metastasis by compounds isolated from green and black teas. Kumagai et al, JP 08104628 A2, Apr. 1, 1996 (CA 125: 67741) reports the use of flavones and anthocyanines isolated from Scutellaris baicanlensis roots to inhibit collagenase. Gervasi et al., (1996) Biochem. Biophys. Res. Comm., 228: 530-538, reports the regulation of MMP-2 by some plant lectins and other saccharides. Dubois et al., (1998) FEBS Lett., 427: 275-278, reports the increased secretion of deletorious gelatinase-B (MMP-9) by some plant lectins. Nagase et al., (1998) Planta Med., 64: 216-219, reports the weak inhibition of collagenase (MMPs) by delphinidin, a flavonoid isolated from Solanum melongena.

Other reports discuss the use of extracts to inhibit extracellular proteases. For example, Asano et al., (1998) Immunopharmacology, 39: 117-126, reports the inhibition of TNF-a production using Tripteggium wilfordii Hook F. extracts. Maheu et al., (1998) Arthritis Rheumatol., 41: 81-91, reports the use of avocado/soy bean non-saponifiable extracts in the treatment of arthritis. Makimura et al., (1993). J. Periodontol., 64: 630-636, also reports the use of green tea extracts to inhibit collagenases in vitro. Obayashi et al., (1998) Nippon Keshonin Gijutsusha Kaishi, 32: 272-279 (CA 130: 92196) reports the inhibition of collagenase-I (MMP-1) from human fibroblast and neutrophil elastase by plant extract from Eucalyptus and Elder.

When a plant is stressed, several biochemical processes are activated and many new chemicals, in addition to those constitutively expressed, are synthesised as a response. These chemicals include enzymes, enzyme inhibitors (especially protease inhibitors), lectins, alkaloids, terpenes, oligosaccharides, and antibiotics. The biosynthesis of these defense chemicals and secondary metabolites is not yet fully understood. The most studied system is the production of protease inhibitors following pest attack or mechanical wounding. On the other hand, several inducible chemicals are the products of complex biochemical pathways which require several biosynthetic enzymes to be activated.

It has been shown that many chemicals can be used to “stress” plants and to artificially stimulate biosynthesis of several new and constitutive defense chemicals. Also, different types of stress can activate distinct metabolic defense pathways, thereby leading to production of a variety of chemicals. Although the various biosynthetic defense pathways share some similarities, these pathways are characteristic of specific plant species. Therefore, treating many plants with many types of stress can lead to a vast number of collections of diverse chemicals from plant origin.

In addition to pests, fungi, and other pathogenic attacks, stressors include drought, heat, water and mechanical wounding. Furthermore, many chemicals can act as stressors that activate gene expression; these include: hydrogen peroxide, ozone, sodium chloride, jasmonic acid and derivatives, α-linoleic acid, ‘γ-linoleic acid, salicylic acid, abscesic acid, volicitin, small oligopeptides, among others.

The use of abiotic stressors on plants has been the focus of intense studies in plant science. Artificial stresses have been used to stimulate the production of natural plant protease inhibitors for insect digestive proteases, in order to enhance crop protection against certain pests and herbivores. They have proven useful in combination with plants genetically modified to express other protease inhibitor genes. Finally, in the area of molecular farming, stresses have been used to stimulate gene expression in plants genetically modified to include an inducible coding sequence for a protein of nutraceutical and/or medicinal interest (Ryan and Farmer, U.S. Pat. No. 5,935,809).

Likewise, the use of gene activators or elicitors have been described to enhance the production of volatile chemicals in plant cell cultures. These elicitors have been demonstrated to induce the activity of several enzymes such as for example phenylalanine ammonia lyase, therefore leading to an increase in the production of plant volatile components.

No one has used stress to improve or modify plants human protease inhibitor content.

BRIEF DESCRIPTION OF THE FIGURES AND TABLES

FIG. 1 presents an overview of one standard procedure that is followed in order to generate the extracts of the invention each of which is derived from the solid plant material. Solvent A, B and C generally represent separate classes of solvents, for example, aqueous, alcoholic and organic. They are generally applied in a polar to non-polar order. They can be applied in a non-polar to polar order, however, in each case the solid matter must be dried prior to contacting the solid matter with the subsequent solvent.

FIG. 2 describes in further detail, one standard procedure that is followed in order to generate the extracts of the invention.

FIG. 3 presents an overview of one example of a commercial procedure that could be followed to prepare extracts of the invention.

Table 1 reports the inhibition of human MMP-1 by aqueous (A), ethanolic (R) and organic (S) 25 extracts for exemplary stressed and non-stressed plant sources.

Table 2 reports the inhibition of human MMP-2 by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed and non-stressed plant sources.

Table 4 reports the inhibition of human MMP-9 by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed and non-stressed plant sources.

Table 5 reports the inhibition of human Cathepsin B by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed and non-stressed plant sources.

Table 6 reports the inhibition of human Cathepsin D by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed and non-stressed plant sources.

Table 7 reports the inhibition of human Cathepsin G by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed and non-stressed plant sources.

Table 8 reports the inhibition of human Cathepsin L by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed and non-stressed plant sources.

Table 9 reports the inhibition of human Cathepsin K by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed and non-stressed plant sources.

Table 10 reports the inhibition of FILE by aqueous (A), ethanolic (R) and organic (S) extracts for 20 exemplary stressed and non-stressed plant sources.

Table 11 reports the inhibition of bacteria Clostripain by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed and non-stressed plant sources.

Table 12 reports the inhibition of bacteria subtilisin by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed and non-stressed plant sources.

SUMMARY OF THE INVENTION

In one aspect the invention provides n extract from a plant, which inhibits the activity of one or more extracellular proteases, wherein the extract has been prepared by the steps of harvesting plant material, treating plant material with a solvent, separating the resulting extract from the solid material. testing an aliquot of the extract against a panel of extracellular proteases, and retaining the extract if it inhibits the activity of one or more extracellular proteases. an extract.

In one aspect the invention provides a library of extracts from plants wherein each extract inhibits the activity of one or more extracellular proteases.

In another aspect the invention provides a library of plant extracts formed by a process comprising: contacting plant material with either an aqueous, ethanolic, or an organic solvent; isolating an extract from said plant material; analysing said extract for the presence of one or more inhibitory activities against an extracellular protease; and collected together, so as to form a library of plant extracts wherein each extract inhibits one or more extracellular proteases.

In one aspect the invention provides an extract from a plant, which inhibits the activity of one or more extracellular proteases, wherein said plant has been stressed prior to generating the extract.

In a further aspect the invention provides a library of extracts derived from plants wherein each extract inhibits the activity of one or more extracellular proteases and wherein said 20 plants have been stressed prior to generating the extract.

In yet a further aspect provides an extracellular protease inhibitor derived from a plant comprising the steps of: contacting plant material with either an aqueous, ethanolic, or an organic solvent; isolating an extract from said plant material; analysing said extract for the presence of one or more inhibitory activities against a panel of extracellular proteases; further purifying a compound from said extract if said extract demonstrates the inhibition of one or more extracellular proteases greater than about 20%.

In another aspect the invention provides a method for increasing the levels of extracellular 30 protease inhibitors in plants comprising the step of stressing the plant prior to forming a plant extract.

In another aspect the invention provides for the use of such extracts during protein purification to minimize the degradation due to extracellular proteases.

DETAILED DESCRIPTION OF THE INVENTION Definitions

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.

“Extracellular protease” means enzymes which degrade proteins (proteases) secreted outside the cell. Included MMPs, cathepsins, elastase, plasmin, TPA, uPA, kallikrein, ADAMS family members, neprilysin, gingipain, clostripain, thermolysin, serralysin, and other bacterial and viral enzymes.

“Extract of the invention,” means a composition prepared by contacting a solvent with plant material, produced following the procedures of the invention, which demonstrates inhibitory activity against one or more extracellular proteases. In one embodiment an extract of the invention demonstrates inhibitory activity against two or more extracellular proteases. In one embodiment an extract of the invention demonstrates inhibitory activity against three or more extracellular proteases. In one embodiment, an extract of the invention demonstrates inhibitory activity against four or more extracellular proteases. The solvent may be evaporated leaving a solid embodiment of the extract. In one embodiment, the inhibitory activity is greater than about 20% when measured according to one of the assays as described herein. In one embodiment a panel of extracellular proteases can be used to test the inhibitory activity of the extract.

“Panel of Extracellular Proteases” means the array of distinct extracellular proteases that are used to perform routine assays to monitor the presence or absence of inhibitory activity throughout the extraction process of the invention. In one embodiment, inhibitory activity against one or more extracellular proteases is monitored; in one embodiment, inhibitory activity against two or more extracellular proteases is monitored; in one embodiment inhibitory activity against three or more extracellular proteases is monitored; in one embodiment inhibitory activity against four or more extracellular proteases is monitored; in one embodiment inhibitory activity against five or more extracellular proteases is monitored. One skilled in the art would appreciate that as high throughput screening techniques develop, one could routinely assay the fractions of the extracts with as many extracellular proteases as the technology permits. In general, the more enzymes that can be routinely tested the more information that can be generated during this process that will be useful for defining extracts useful to inhibit extracellular proteases.

“Potential plants” includes all species of the Kingdom Plantae, including plants under the Division Chlorophyta, Division Rhodophora, Division Paeophyta, Division Bryophyta and Division Tracheophyta; Subdivision Lycopsida, Subdivision Sphenopsida, Subdivision Pteropsida and Subdivision Spermopsida; Class Gymnospermae, Class Angiospermae, Subclass Dicotyledonidae and Subclass Monocotyledonidae. In general terms, all plants, herbs, and lower plants such as fungi and algae. Potential plants are those plants that can be subjected to the methodology of the invention in order to generate an extract which can then be tested against a panel of extracellular proteases. Those plants which yield an extract demonstrating inhibitory activity against an extracellular protease are considered to be plants and extracts comprising the subject matter of the invention.

“Potential Pre-Extract” means an extract which has not yet been determined to posess inhibitory activity against one or more extracellular proteases.

“Plant material” means any part of a plant taken individually or in group, could include but not restricted to leafs, flowers, roots, seeds, stems, and other part of a plant, wherein a plant may be terrestrial, aquatic or other.

“Protease inhibitor” as used herein, refers to any compound that attenuates the proteolytic activity of proteases. “Protease inhibitor” may or may not be proteinaceous.

“Stressor” as used herein, refers to any physical stress, chemical compound, or a biological agent used to elicit production of extracellular protease inhibitors as a result of activation of a defence response in a plant. Elicitors and inducers are also considered to be stressors. Any material of a plant may be contacted with a stressor, elicitor, or inducer, which is a chemical compound, for example organic and inorganic acids, fatty acids, glycerides, phopholipids, glycolipids, orgnaic solvents, amino acids, and peptides, monosaccharides, oligosaccharides, polysaccharides and lipopllysaccharides, phenolics, alkaloids, terpenes and terpenoids, antibiotics, detergents, polyamines, peroxides, ionophores, etc., or subjected to a physical treatment, such as ultraviolet radiation, low and high temperature stress, osmotic stress induced by salt or sugars, nutritional stress defined as depriving the plant of essential nutrients (N, P, or K), in order to induce or elicit increased production of one or more chemicals. Such chemical compound or physical treatment may be applied continuously or intermittently to the plant or plant part. In one embodiment, such treatment may be accomplished by contacting the plant material with a solution containing the elicitor or by irradiating the plant material or exposing the plant material to other environmental stresses such as temperature stresses.

The term “substantially purified” or “substantially pure” or “isolated,” when used in reference to a molecule having protease inhibitor activity, means that the molecule is in a form that is relatively free of proteins, nucleic acids, lipids, carbohydrates or other materials with which it is naturally associated in a plant. As disclosed herein, a plant extract of the invention is considered to be substantially purified. In addition, the molecules having protease inhibitor activity can be further purified using routine and well known methods as provided herein. As such, a substantially pure protease inhibitor of the invention can constitute at least about one or a few percent of a sample, for example, at least about five percent of a sample, generally at least about twenty percent of a sample, and can be further purified to constitute at least about fifty percent of a sample, generally at least about eighty percent of a sample, and particularly about ninety percent or ninety-five percent or more of a sample. A determination that a protease inhibitor of the invention is substantially pure can be made using methods as disclosed herein or otherwise known in the art, for example, by performing electrophoresis and identifying the particular molecule as a relatively discrete band.

Other chemistry terms herein are used according to conventional usage in the art, as exemplified by The McGraw-Hill Dictionary of Chemical Terms (ed. Parker, S., 1985), 30 McGraw-Hill, San Francisco, incorporated herein by reference).

The subject invention involves extracts from the tissues of plant species which provide inhibitory activity against extracellular proteases. In one embodiment, the present invention relates to the use of plants to produce extracts or semi-purified/purified compounds, compositions and formulations demonstrating an inhibitory activity against one or more proteases involved in the proteolytic degradation of human extracellular matrix. Such extracts, compounds, compositions and formulations derived from plant sources, optionally from water, ethanol or organic extracts prepared from said plant tissues, and fractions separable from said extracts by chromatography or centrifugal ultra-filtration or other means. In one aspect, these extracts with inhibitory activity can be used during protein purification to minimize the degradation due to extracellular proteases.

With reference to FIG. 1, the process for producing an extract of the invention begins with choosing a plant species. Then a pre-harvest treatment is selected, wherein either treatment with water, or water in addition to any combination of a stress, wherein the stress can be applied separately from the water (if the stress is drought, then the water would not be provided for the period in which the plant is to be stressed); followed by choosing whether the treated plant will be treated for storage and stored prior to contacting plant material with the first solvent. The plant material is treated with the first solvent and then the liquid is separated from the solid material (solid S2), wherein the liquid becomes Fraction F1 or Pre-Extract A. The solid S2 is treated with the second solvent and then the liquid is separated from the solid material (Solid S3), wherein the liquic becomes Fraction F2 or Pre-Extract B. The solid S3 is treated with the third solvent and then the liquid is separated from the solid material (Solid S4).

Plant Material

In one embodiment, plants that may be employed in the invention comprise: Abelmoschus esculentus; Abies balsamea; Abies lasiocarpa; Achillea millefolium; Achillea tomentosa; Aconitum napellus; Aconitum spp.; Acorus calamus; Actaea racemosa; Actinidia arguta; Actinidia chinensis; Adiantum pedatum; Adiantum tenerum; Aesculus hippocastanum; Aframomum melegueta; Agaricus bisporus; Agastache foeniculum; Ageratum conyzoides; Agrimonia eupatoria; Agropyron cristatum; Agropyron repens; Agrostis alba; Agrostis stolonifera; Alcea rosea; Alchemilla twills; Alkanna tinctoria; Allium ampeloprasum; Allium cepa; Allium fistulosum; Allium grande; Allium porrum; Allium sativum; Allium schoenoprasum; Allium tuberosum; Allium victorialis; Aloe vera; Alpinia officinarum; Althaea officinalis; Amaranthus caudatus; Amaranthus retroflexus; Amaranthus tricolor; Ambrosia artemisiifolia; Amelanchier alnifolia; Amelanchier canadensis; Amelanchier sanguinea; Amelanchier sanguinea×A. laevis; Amsonia tabemaemontana; Ananas comosus; Anaphalis margaritacea; Anethum graveolens; Angelica archangelica; Angelica dahurica; Angelica sinensis; Anthemis tinctoria; Anthoxanthum odoratum; Anthriscus cerefolium; Anthurium guildingii; Apium graveolens; Apocynum cannabinum; Arachis hypogaea; Aralia cordata; Aralia nudicaulis; Arctium lappa; Arctium minus; Arctostaphylos uva-ursi; Armoracia rusticana; Aronia melanocarpa; Aronia×prunifolia; Arrhenatherum elatius; Artemisia abrotanum; Artemisia absinthium; Artemisia dracunculus; Artemisia ludoviciana; Artemisia vulgaris; Asarum europaeum; Asclepias incarnata; Asclepias tuberosa; Asparagus officinalis; Aster spp.; Astilbe×arendsii; Astilboides tabularis; Athyrium asperum; Atriplex hortensis; Atropa belladonna; Avena sativa; Averrhoa carambola; Baptisia tinctoria; Beckmannia eruciformis; Begonia convolvulacea; Begonia eminii; Begonia glabra; Begonia mannii; Begonia polygonoides; Bellis perennis; Berberis vulgaris; Beta vulgaris; Betula alleghaniensis; Betula glandulosa; Boesenbergia rotunda; Boletus edulis; Borago officinalis; Brassica cepticepa; Brassica juncea; Brassica napus; Brassica nigra; Brassica oleracea; Brassica rapa; Bromus inermis; Buddleja davidii; Bupleurum falcatum; Butomus umbellatus; Caladium spp.; Calamagrostis arundiflora; Calamintha nepeta; Calendula officinalis; Camellia sinensis; Campanula rapunculus; Canna indica; Cantharellus cibarius; Capsella bursa-pastoris; Capsicum annuum; Capsicum frutescens; Carex morrowii; Carica papaya; Carthamus tinctorius; Carum carvi; Carya cordiformis; Castanea spp.; Centaurea solstitialis; Cerastium tomentosum; Chaerophyllum bulbosum; Chamaemelum nobile; Chelidonium majus; Chenopodium album; Chenopodium bonus-henricus; Chenopodium quinoa; Chrysanthemum coronarium; Cicer arietinum; Cichorium endivia subsp. endivia; Cichorium intybus; Cinnamomum verum; Cirsium arvense; Cissus discolor; Citrullus colocynthis; Citrullus lanatus; Citrus limettoides; Citrus limon; Citrus reticulata; Citrus sinensis; Citrus×paradisi; Clematis armandii; Clematis chiisanensis; Coccoloba caracasana; Cocos nucifera; Coix lacrymajobi; Colocasia spp.; Convallaria majalis; Conyza canadensis; Corchorus olitorius; Coriandrum sativum; Cornus canadensis; Cornus mas; Cosmos sulphureus; Cotinus coggygria; Crataegus sanguinea; Crataegus spp.; Crataegus submollis; Crithmum maritimum; Cryptotaenia canadensis; Cucumis anguria; Cucumis melo; Cucumis metuliferus; Cucumis sativus; Cucurbita maxima; Cucurbita moschata; Cucurbita pepo; Cullen corylifolium; Cuminum cyminum; Curcuma longa; Curcuma zedoaria; Cyclonia oblonga; Cymbopogon citratus; Cymbopogon martinii; Cynara cardunculus subsp. cardunculus; Cyperus esculentus; Dactylis glomerata; Datisca cannabina; Datura metel; Datura stramonium; Daucus carota; Digitalis purpurea; Dimocarpus longan; Dioscorea batatas; Diospyros kaki; Dipsacus sativus; Dirca palustris; Dolichos lablab; Dryopteris filix-mas; Echinacea purpurea; Echinochloa frumentacea; Eleusine coracana; Equisetum hyemale; Erigeron speciosus; Eriobotrya japonica; Eruca vesicaria; Erysimum perofskianum; Eschscholzia californica; Fagopyrum esculentum; Fagopyrum tataricum; Festuca rubra; Filipendula rubra; Filipendula ulmaria; Filipendula vulgaris; Foeniculum vulgare; Forsythia×intermedia; Fortunella spp.; Fragaria×ananassa; Frangula alnus; Fucus vesiculosus; Fumaria officinalis; Galinsoga quadriradiata; Galium odoratum; Gaultheria hispidula; Gaultheria procumbens; Genista multibracteata; Gentiana lutea; Gentiana macrophylla; Geum rivale; Ginkgo biloba; Glechoma hederacea; Glyceria maxima; Glycine max; Glycyrrhiza glabra; Gossypium herbaceum; Guizotia abyssinica; Hamamelis virginiana; Hedeoma pulegioides; Hedychium spp.; Helianthus annuus; Helianthus strumosus; Helianthus tuberosus; Helichrysum angustifolium; Helichrysum thianschanicum; Heliotropium arborescens; Helleborus niger; Herba schizonepetae; Hibiscus cannabinus; Hordeum hexastichon; Hordeum vulgare; Hordeum vulgare subsp. vulgare; Houttuynia cordata; Humulus lupulus; Hydrastis canadensis; Hylotelephium spp.; Hymenoxys hoopesii; Hyoscyamus niger; Hypericum henryi; Hypericum perforatum; Hypericum spp.; Hypomyces lactifluorum; Hyssopus officinalis; Iberis amara; Iberis sempervirens; Inula helenium; Ipomoea batatas; Iris versicolor; Isatis tinctoria; Jeffersonia diphylla; Juglans nigra; Juniperus communis; Kochia scoparia; Koeleria glauca; Kolkwitzia amabilis; Krameria lappacea; Lactuca sativa; Lactuca serriola; Laportea canadensis; Laserpitium latifolium; Lathyrus sativus; Lathyrus sylvestris; Laurus nobilis; Lavandula angustifolia; Lavandula latifolia; Ledum groenlandicum; Lens culinaris subsp. culinaris; Lentinus edodes; Leonurus cardiaca; Lepidium sativum; Leucanthemum vulgare; Levisticum officinale; Ligularia dentata; Ligustrum vulgare; Linaria vulgaris; Lindera benzoin; Linum usitatissimum; Litchi chinensis; Lolium multiflorum; Lolium perenne; Lonicera ramosissima; Lonicera syringantha; Lotus corniculatus; Lotus tetragonolobus; Lunaria annua; Lupinus polyphyllus; Luzula sylvatica; Lychnis chalcedonica; Lycopersicon esculentum; Lycopersicon pimpinellifolium; Lysimachia clethroides; Lythrum salicaria; Madia sativa; Magnolia stellata; Malus hupehensis; Malus prunifolia; Malus spp.; Malva moschata; Malva sylvestris; Mangifera indica; Manihot esculenta; Marrubium vulare; Matricaria recutita; Matricaria spp.; Medicago sativa; Melaleuca alternifolia; Melilotus albus; Melilotus officinalis; Melissa officinalis; Mentha arvensis; Mentha pulegium; Mentha spicata; Mentha suaveolens; Mentha×piperita; Menyanthes trifoliata; Microlepia platyphylla; Miscanthus sacchariflorus; Miscanthus sinensis; Momordica charantia; Monarda didyma; Monarda fistulosa; Monarda spp.; Musa×paradisiaca; Myrica pensylvanica; Nasturtium officinale; Nepeta cataria; Nicotiana rustica; Nicotiana tabacum; Nigella sativa; Ocimum Basilicum; Oenothera biennis; Onobrychis viciifolia; Ophiopogon japonicus; Opuntia spp.; Origanum majorana; Origanum vulgare; Oryza sativa; Oxalis deppei; Oxyria digyna; Paeonia rubra; Paeonia spp.; Panax quinquefolius; Panicum miliaceum; Passiflora caerulea; Passiflora spp.; Pastinaca sativa; Pennisetum alopecuroides; Perilla frutescens; Persea americana; Petasites japonicus; Petroselinum crispum; Peucedanum cervaria; Peucedanum oreaselinum; Pfaffia paniculata; Phacelia tanacetifolia; Phalaris arundinacea; Phalaris canariensis; Phaseolus acutifolius; Phaseolus coccineus; Phaseolus vulgaris; Philadelphus coronarius; Phleum pratense; Phlox paniculata; Phoenix dactylifera; Physalis grisea; Physalis philadelphica; Physalis spp.; Physostegia virginiana; Phytolacca americana; Pimpinella anisum; Pisum sativum; Plantago coronopus; Plantago major; Plectranthus fruticosus; Plectranthus spp.; Pleurotus spp.; Plumbago zeylanica; Poa compressa; Poa pratensis; Podophyllum peltatum; Polygonatum odoratum; Polygonum aviculare; Polygonum chinense; Polygonum pensylvanicum; Polygonum persicaria; Pongamia pinnata; Pontederia cordata; Populus incrassata; Populus tremula; Populus×petrowskyana; Portulaca oleracea; Potentilla anserina; Poterium sanguisorba; Primula veris; Prunella vulgaris; Prunus armeniaca; Prunus cerasus; Prunus persica; Prunus spp.; Prunus tomentosa; Psathyrostachys juncea; Psidium guajava; Psidium spp.; Pteridium aquilinum; Pulmonaria officinalis; Pulmonaria saccharata; Punica granatum; Pyrus communis; Pyrus pyrifolia; Raphanus raphanistrum; Raphanus sativus; Rehmannia glutinosa; Reseda luteola; Reseda odorata; Rheum officinale; Rheum palmatum; Rheum×hybridum; Rhus aromatica; Rhus trilobata; Ribes grossularia; Ribes nigrum; Ribes rubrum; Ribes sylvestre; Ribes uva-crispa; Ribes×nidigrolaria; Ricinus communis; Rosa rugosa; Rosmarinus officinalis; Rubus allegheniensis; Rubus canadensis; Rubus idaeus; Rubus occidentalis; Rubus thibetanus; Rumex acetosa; Rumex acetosella; Rumex crispus; Rumex patientia; Rumex scutatus; Ruta graveolens; Saccharum officinarum; Salix purpurea; Salvia elegans; Salvia officinalis; Salvia sclarea; Salvia sylvestris; Sambucus canadensis; Sambucus ebulus; Sambucus nigra; Sanguisorba minor; Sanguisorba officinalis; Santolina chamaecyparissus; Saponaria officinalis; Satureja hortensis; Satureja montana; Satureja repandra; Scolymus hispanicus; Scorzonera hispanica; Scrophularia nodosa; Scutellaria lateriflora; Secale cereale; Sechium edule; Senecio vulgaris; Serenoa repens; Serratula tinctoria; Sesamum indicum; Setaria italica; Sidalcea spp.; Silene vulgaris; Silybum marianum; Sinapis alba subsp. alba; Sium sisarum; Solanum dulcamara; Solanum melongena; Solanum scabrum; Solanum tuberosum; Solidago canadensis; Solidago spp.; Solidago virgaurea; Solidago×hybrida; Sonchus oleraceus; Sorghum bicolor; Sorghum×drummondii; Spinacia oleracea; Stachys affinis; Stachys byzantina; Stachys macrantha; Stellaria graminea; Stellaria media; Stipa capillata; Symphytum officinale; Tamarindus indica; Tanacetum balsamita; Tanacetum balsamita subsp. balsamita; Tanacetum cinerariifolium; Tanacetum parthenium; Tanacetum vulgare; Taraxacum officinale; Tetradenia riparia; Teucrium chamaedrys; Thalictrum aquilegiifolium; Thlaspi arvense; Thuja occidentalis; Thymus fragantissimus; Thymus herba-barona; Thymus praecox subsp. arcticus; Thymus pseudolanuginosus; Thymus serpyllum; Thymus vulgaris; Thymus×citriodorus; Tiarella cordifolia; Tiarella spp.; Tragopogon porrifolius; Tragopogon spp.; Trichosanthes kirilowii; Trifolium hybridum; Trifolium incamatum; Trifolium pannonicum; Trifolium pratense; Trifolium repens; Trigonella foenum-graecum; Triticum aestivum; Triticum aestivum subsp. spelta; Triticum turgidum; Trollius×cultorum; Tropaeolum majus; Tsuga canadensis; Tsuga diversifolia; Tsuga mertensiana; Tussilago farfara; Typha latifolia; Ulmus americana; Urtica dioica; Uvularia perfoliata; Vaccinium angustifolium; Vaccinium corymbosum; Vaccinium macrocarpon; Valeriana officinalis; Valerianella locusta; Veratrum viride; Verbascum thapsus; Verbena officinalis; Veronica officinalis; Viburnum opulus; Vicia faba; Vicia sativa; Vicia villosa; Vigna angularis; Vigna mungo; Vigna unguiculata; Vinca minor; Vitis spp.; Weigela coraeensis; Weigela hortensis; Withania somnifera; ×Triticosecale spp.; Xanthium sibiricum; Xanthium strumarium; Yucca filamentosa; Zea mays; Zingiber officinale; Achillea ptarmica; Ajuga reptans; Aster spp; Astilbe chinensis; Bergenia×schmidtii; Brassica chinensis; Butomus umbellatus; Buxus microphylla; Carpinus caroliniana; Centaurea dealbata; Chaenomeles×superba; Clematis alpina; Coreopsis verticillata; Corpus alba; Corpus sericea; Corylus maxima; Crambe cordifolia; Cyperus alternifolius; Dahlia spp.; Euphorbia amygdaloides; Fuchsia spp.; Fuchsia magellanica; Galium aparine; Geranium sanguineum; Geranium phaeum; Geranium pratense; Geranium sanguineum; Geranium×cantabrigiense; Glaux Maritima; Hamamelis mollis; Hedychium coronarium; Helenium spp.; Herba Schizonepetae; Hosta sieboldiana; Hydrangea quercifolia; Ipomoea aquatica; Lamiastrum galeobdolon; Magnolia×loebneri; Malva verticillata; Matteuccia pensylvanica; Microbiata decussata; Montia perfoliata; Ocimum tenuiflorum; Oenothera fruticosa subsp fruticosa; Onoclea sensibilis; paeonia suffruticosa; Penstemon digitalis; Petasites japonicus; Physalis alkekengi; Pinus cembra; Pinus mugo; Potentilla fruticosa; Rhododendron spp.; ribes americanum; Rodgersia spp.; Rodgersia podophylla; Rubus arcticus; Rubus phoenicolasius; Rubus pubescens; Rudbeckia maxima; Sempervivum tectorum; Soleirolia soleirolii; Solidago caesia; Staphylea trifolia; Stephanandra incisa; Stewartia pseudocamellia; Strelitzia reginae; Symphoricarpos orbiculatus; Symphoricarpos albus; Taxus×media; Vernonia gigantea; Veronica austriaca ssp teucrium; Veronica beccabunga; Viburnum plicatum.

It is further contemplated by this invention that any plant may be employed in the method as a potential plant. For example, plants belonging to the following classifications may optionally be employed in order to prepare an extract of the invention when such extracts are demonstrated to possess inhibitory activities against extracellular proteases: Superdivision Spermatophyta—Seed plants Division Coniferophyta—Conifers Class Pinopsida Order Finales Family Araucariaceae Araucaria family Family Cephalotaxaceae—Plum Yew family Family Cupressaceae—Cypress family Family Pinaceae—Pine family Family Podocarpaceae—Podocarpus family Family Taxodiaceae—Redwood family Order Taxales Family Taxaceae—Yew family Division Cycadophyta—Cycads Class Cycadopsida Order Cycadales Family Cycadaceae—Cycad family Family Zamiaceae—Sago-palm family Division Ginkgophyta—Ginkgo Class Ginkgoopsida Order Ginkgoales Family Ginkgoaceae—Ginkgo family Division Gnetophyta—Mormon tea and other gnetophytes Class Gnetopsida Order Ephedrales Family Ephedraceae—Mormon-tea family Order Gnetales Family Gnetaceae Gnetum family Division Magnoliophyta—Flowering plants Class Liliopsida—Monocotyledons Subclass Alismatidae Order Alismatales Family Alismataceae—Water-plantain family Family Butomaceae—Flowering Rush family Family Limnocharitaceae—Water-poppy family Order Hydrocharitales Family Hydrocharitaceae—Tape-grass family Order Najadales Family Aponogetonaceae—Cape-pondweed family Family Cymodoceaceae—Manatee-grass family Family Juncaginaceae—Arrow-grass family Family Najadaceae—Water-nymph family Family Posidoniaceae—Posidonia family Family Potamogetonaceae—Pondweed family Family Ruppiaceae—Ditch-grass family Family Scheuchzeriaceae—Scheuchzeria family Family Zannichelliaceae—Horned pondweed family Family Zosteraceae—Eel-grass family Subclass Arecidae Order Arales Family Acoraceae—Calamus family Family Araceae—Arum family Family Lemnaceae—Duckweed family Order Arecales Family Arecaceae—Palm family Order Cyclanthales Family Cyclanthaceae—Panama Hat family Order Pandanales Family Pandanaceae—Screw-pine family Subclass Commelimidae Order Commelinales Family Commelinaceae—Spiderwort family Family Mayacaceae—Mayaca family Family Xyridaceae—Yellow-eyed Grass family Order Cyperales Family Cyperaceae—Sedge family Family Poaceae—Grass family Order Eriocaulales Family Eriocaulaceae—Pipewort family Order Juncales Family Juncaceae—Rush family Order Restionales Family Joinvilleaceae—Joinvillea family Order Typhales Family Sparganiaceae—Bur-reed family Family Typhaceae—Cat-tail family Subclass Liliidae Order Liliales Family Agavaceae—Century-plant family Family Aloeaceae—Aloe family Family Dioscoreaceae—Yam family Family Haemodoraceae—Bloodwort family Family Hanguanaceae—Hanguana family Family Iridaceae—Iris family Family Liliaceae—Lily family Family Philydraceae—Philydraceae family Family Pontederiaceae—Water-Hyacinth family Family Smilacaceae—Catbrier family Family Stemonaceae—Stemona family Family Taccaceae—Tacca family Order Orchidales Family Burmanniaceae—Burmannia family Family Orchidaceae—Orchid family Subclass Zingiberidae Order Bromeliales Family Bromeliaceae—Bromeliad family Order Zingiberales Family Cannaceae—Canna family Family Costaceae—Costus family Family Heliconiaceae—Heliconia family Family Marantaceae—Prayer-Plant family Family Musaceae—Banana family Family Zingiberaceae—Ginger family Class Magnoliopsida—Dicotyledons Subclass Asteridae Order Asterales Family Asteraceae—Aster family Order Callitrichales Family Callitrichaceae—Water-starwort family Family Hippuridaceae—Mare's-tail family Order Calycerales Family Calyceraceae—Calycera family Order Campanulales Family Campanulaceae—Bellflower family Family Goodeniaceae—Goodenia family Family Sphenocleaceae—Spenoclea family Order Dipsacales Family Adoxaceae—Moschatel family Family Caprifoliaceae—Honeysuckle family Family Dipsacaceae—Teasel family Family Valerianaceae—Valerian family Order Gentianales Family Apocynaceae—Dogbane family Family Asclepiadaceae—Milkweed family Family Gentianaceae—Gentian family Family Loganiaceae—Logania family Order Lamiales Family Boraginaceae—Borage family Family Lamiaceae—Mint family Family Lennoaceae—Lennoa family Family Verbenaceae—Verbena family Order Plantaginales Family Plantaginaceae—Plantain family Order Rubiales Family Rubiaceae—Madder family Order Scrophulariales Family Acanthaceae—Acanthus family Family Bignoniaceae—Trumpet-creeper family Family Buddlejaceae—Butterfly-bush family Family Gesneriaceae—Gesneriad family Family Lentibulariaceae—Bladderwort family Family Myoporaceae—Myoporum family Family Oleaceae—Olive family Family Orobanchaceae—Broom-rape family Family Pedaliaceae—Sesame family Family Scrophulariaceae—Figwort family Order Solanales Family Convolvulaceae—Morning-glory family Family Cuscutaceae—Dodder family Family Fouquieriaceae—Ocotillo family Family Hydrophyllaceae—Waterleaf family Family Menyanthaceae—Buckbean family Family Polemoniaceae—Phlox family Family Solanaceae—Potato family Subclass Caryophyllidae Order Caryophyllales Family Achatocarpaceae—Achatocarpus family Family Aizoaceae—Fig-marigold family Family Amaranthaceae—Amaranth family Family Basellaceae—Basella family Family Cactaceae—Cactus family Family Caryophyllaceae—Pink family Family Chenopodiaceae—Goosefoot family Family Molluginaceae—Carpet-weed family Family Nyctaginaceae—Four o'clock family Family Phytolaccaceae—Pokeweed family Family Portulacaceae—Purslane family Order Plumbaginales Family Plumbaginaceae—Leadwort family Order Polygonales Family Polygonaceae—Buckwheat family Subclass Dilleniidae Order Batales Family Bataceae—Saltwort family Order Capparales Family Brassicaceae—Mustard family Family Capparaceae—Caper family Family Moringaceae—Horse-radish tree family Family Resedaceae—Mignonette family Order Diapensiales Family Diapensiaceae—Diapensia family Order Dilleniales Family Dilleniaceae—Dillenia family Family Paeoniaceae—Peony family Order Ebenales Family Ebenaceae—Ebony family Family Sapotaceae—Sapodilla family Family Styracaceae—Storax family Family Symplocaceae—Sweetleaf family Order Ericales Family Clethraceae—Clethra family Family Cyrillaceae—Cyrilla family Family Empetraceae—Crowberry family Family Epacridaceae—Epacris family Family Ericaceae—Heath family Family Monotropaceae—Indian Pipe family Family Pyrolaceae—Shinleaf family Order Lecythidales Family Lecythidaceae—Brazil-nut family Order Malvales Family Bombacaceae—Kapok-tree family Family Elaeocarpaceae—Elaeocarpus family Family Malvaceae—Mallow family Family Sterculiaceae—Cacao family Family Tiliaceae—Linden family Order Nepenthales Family Droseraceae—Sundew family Family Nepenthaceae—East Indian Pitcher-plant family Family Sarraceniaceae—Pitcher-plant family Order Primulales Family Myrsinaceae—Myrsine family Family Primulaceae—Primrose family Family Theophrastaceae—Theophrasta family Order Salicales Family Salicaceae—Willow family Order Theales Family Actinidiaceae—Chinese Gooseberry family Family Caryocaraceae—Souari family Family Clusiaceae—Mangosteen family Family Dipterocarpaceae—Meranti family Family Elatinaceae—Waterwort family Family Marcgraviaceae—Shingle Plant family Family Ochnaceae—Ochna family Family Theaceae—Tea family Order Violales Family Begoniaceae—Begonia family Family Bixaceae—Lipstick-tree family Family Caricaceae—Papaya family Family Cistaceae—Rock-rose family Family Cucurbitaceae—Cucumber family Family Datiscaceae—Datisca family Family Flacourtiaceae—Flacourtia family Family Frankeniaceae—Frankenia family Family Loasaceae—Loasa family Family Passifloraceae—Passion-flower family Family Tamaricaceae—Tamarix family Family Tumeraceae—Turnera family Family Violaceae—Violet family Subclass Hamamelidae Order Casuarinales Family Casuarinaceae—She-oak family Order Fagales Family Betulaceae—Birch family Family Fagaceae—Beech family Order Hamamelidales Family Cercidiphyllaceae—Katsura-tree family Family Hamamelidaceae—Witch-hazel family Family Platanaceae—Plane-tree family Order Juglandales Family Juglandaceae—Walnut family Order Leitneriales Family Leitneriaceae—Corkwood family Order Myricales Family Myricaceae—Bayberry family Order Urticales Family Cannabaceae—Hemp family Family Cecropiaceae—Cecropia family Family Moraceae—Mulberry family Family Ulmaceae—Elm family Family Urticaceae—Nettle family Subclass Magnoliidae Order Aristolochiales Family Aristolochiaceae—Birthwort family Order Illiciales Family Illiciaceae—Star-anise family Family Schisandraceae—Schisandra family Order Laurales Family Calycanthaceae—Strawberry-shrub family Family Hernandiaceae Hernandia family Family Lauraceae—Laurel family Family Monimiaceae—Monimia family Order Magnoliales Family Annonaceae—Custard-apple family Family Canellaceae—Canella family Family Magnoliaceae—Magnolia family Family Myristicaceae—Nutmeg family Family Sonneratiaceae—Sonneratia family Family Winteraceae—Wintera family Order Nymphaeales Family Cabombaceae—Water-shield family Family Ceratophyllaceae—Homwort family Family Nelumbonaceae—Lotus-lily family Family Nymphaeaceae—Water-lily family Order Papaverales Family Fumariaceae—Fumitory family Family Papaveraceae—Poppy family Order Piperales Family Chloranthaceae Chloranthus family Family Piperaceae—Pepper family Family Saururaceae—Lizard's-tail family Order Ranunculales Family Berberidaceae—Barberry family Family Lardizabalaceae—Lardizabala family Family Menispermaceae—Moonseed family Family Ranunculaceae—Buttercup family Family Sabiaceae—Sabia family Subclass Rosidae Order Apiales Family Apiaceae—Carrot family Family Araliaceae—Ginseng family Order Celastrales Family Aquifoliaceae—Holly family Family Celastraceae—Bittersweet family Family Corynocarpaceae—Karaka family Family Hippocrateaceae—Hippocratea family Family Icacinaceae—Icacina family Family Stackhousiaceae—Stackhousia family Order Comales Family Cornaceae—Dogwood family Family Garryaceae—Silk Tassel family Family Nyssaceae—Sour Gum family Order Euphorbiales Family Buxaceae—Boxwood family Family Euphorbiaceae—Spurge family Family Simmondsiaceae—Jojoba family Order Fabales Family Fabaceae—Pea family Order Geraniales Family Balsaminaceae—Touch-me-not family Family Geraniaceae—Geranium family Family Limnanthaceae—Meadow-Foam family Family Oxalidaceae—Wood-Sorrel family Family Tropaeolaceae—Nasturtium family Order Haloragales Family Gunneraceae—Gunnera family Family Haloragaceae—Water Milfoil family Order Linales Family Erythroxyl aceae—Coca family Family Linaceae—Flax family Order Myrtales Family Combretaceae—Indian Almond family Family Lythraceae—Loosestrife family Family Melastomataceae—Melastome family Family Myrtaceae—Myrtle family Family Onagraceae—Evening Primrose family Family Punicaceae—Pomegranate family Family Thymelaeaceae—Mezereum family Family Trapaceae—Water Chestnut family Order Podostemales Family Podostemaceae—River-weed family Order Polygalales Family Krameriaceae—Krameria family Family Malpighiaceae—Barbados Chemy family Family Polygalaceae—Milkwort family Order Proteales Family Proteaceae—Protea family Order Rafflesiales Family Rafflesiaceae—Rafflesia family Order Rhamnales Family Elaeagnaceae—Oleaster family Family Rhamnaceae—Buckthorn family Family Vitaceae—Grape family Order Rhizophorales Family Rhizophoraceae—Red Mangrove family Order Rosales Family Brunelliaceae—Bruneilia family Family Chrysobalanaceae—Cocoa-plum family Family Connaraceae—Cannarus family Family Crassulaceae—Stonecrop family Family Crossosomataceae—Crossosoma family Family Cunoniaceae—Cunonia family Family Grossulariaceae—Currant family Family Hydrangeaceae—Hydrangea family Family Pittosporaceae—Pittosporum family Family Rosaceae—Rose family Family Saxifragaceae—Saxifrage family Family Surianaceae—Suriana family Order Santalales Family Balanophoraceae—Balanophora family Family Eremolepidaceae—Catkin-mistletoe family Family Loranthaceae—Showy Mistletoe family Family Olacaceae—Olax family Family Santalaceae—Sandalwood family Family Viscaceae—Christmas Mistletoe family Order Sapindales Family Aceraceae—Maple family Family Anacardiaceae—Sumac family Family Burseraceae—Frankincense family Family Hippocastanaceae—Horse-chestnut family Family Meliaceae—Mahogany family Family Rutaceae—Rue family Family Sapindaceae—Soapberry family Family Simaroubaceae—Quassia family Family Staphyleaceae—Bladdernut family Family Zygophyllaceae—Creosote-bush family.

In one embodiment, potential plants comprise: Atropa Belladonna, Erythrinia glabeliferus, Ipomea tricolor, Erythrinia crista, Celosia cristata, Gallium sporium, Laurus nobilis, Vitis labrissa, Gratiola officinalis, Symphitium officinalis, Hosta fortuna, Casia hebecarpa, Thalictum flavum, Scutellarian altissima, Portulaca oleacea, Scutellaria certicola, Physalis creticola, Geum fanieri, Gentiana tibetica, Linium hirsutum, Aconitum napellus, Podophyllum amodii, Thymus cretaceus, Hosta fortunaea, Carlina acaulis, Charnaechrista fasciculata, Pinus pinea, Pegamun hamalis, Tamarindus india, Carica papaya, Cistus incanus, Capparis spinosa inemis, Cupress lusitanica, Diopiros kaka, Erungium campestre, Aesculus woerlitzenis, Aesculus hippocastanum, Cupressus sempervirens, Celtis occidentalis, Polygonum cuspidatum, Eleagnus angustifolia, Eleagnus cemutata, Gentiana macrophilla, Brassica napa, Sesbania exaltata, Sesbania speciosa, Spartina potentiflora, Brassica juncea, Helianthus annus, Puansetia sp., Pelargonium zonale, Sundapsis spp., Leontopodium alpinum, Lupinus luteaus, Buxus microphilla “japonica”, Liatris spinata, Rimula japonica, Betula nigra, Filipendula vulgrais, Lobelia siphitica, Gravilia robusta, Reseda luteola, Gentiana littorala, Campanula carpatica, Aesculus hypocastanum, Aesculus waertilensis, Ageratum conizoides, Psidium guajava, Ailantus altissima, Buxus microphylla “japonica”, Hydrocotile asiatica, Gravilea robusta, Brugmansia suaveolens, Thymus puliglodes, Thymus lemabarona, Thymus serphylum (wild), Gaultheria procumbens, Thymus serphylum, Thymus camosus, Thymus thrasicus, Calicatus floridus, Zingiber officinalis, Lapia dulcis, Thymus vulgaris “argenteus”, Thymus praecox “arcticus”, Thymus puleglodes “lemons”, Thymus speciosa, Thymus camosus, Thymus pseudolamginosus, Thymus praecox, Thymus vulgaris “oregano”, Ficus religiosa, Forsithsia suspensa, Chelidonium majus, Thymus wooly, Thymus portugalense, Nicotiana tabacum, Thymus cytridorus “aureus”, Thymus vulgaris, Cactus officinalis, Lal lab purpurea, Juglands regia, Actinidia chinensis, Hernerocalis spp., Betula pendula, Gardenia jasminoides, Taxodium dixticum, Magnolia loebheril, Crataegus praegophyrum, Larix dedidua, Tuja orientalis “eligantissima”, Tula ocidentalis “columbia”, Xeupressocyparis deylandii, Pseudotsuga menzisia, Abies firma, Fautenousus qualiqualia, Alium cernum (wild), Juniperus “blue pacific”, Taraxacum officinalis, Juca sp., Ilex agnifolium, Tsuga canadensis “penola”, Ilex cornuta, Taxus hiksii, Taxus media, Metasequoia glyptotrobioldes, Pinus bungiana, Boxus sempervirens, Stevartia coreana, Prunus xocane, Betula daurica, Plantago minor, Acer palmatum “burgundy”, Acer campestre, Cotynus cogygria, Quercus robur “fastigiata”, Acer truncatum, Archirantus bidentata, Alum japonica, Carum capsicum, Agastache mexuicana, Prunella vulgaris, Tagetes minuta, Nepeta cataria, Ratibiunda columnus-Fera, Aster-Nova anglicae, Mirica certifera, Pittisporum tibica, Taxodium dixticum (H2O), Taxodium dixticum (Acetic acid), Plantago major, Scotch pine, Asorum canadensis, Pieras japonica, Pinus sirtrobus, Trifolium pratense, Prunus serotica, Darura stramonium, Geranium maculata, Hydrocotile asiatica, Astragulus sinicus, Centauria maculata, Ruschia indurata, Myrthus comunis, Platanus acidentalis, Liclum barbatum, Lavandula officinalis, Gravilea robusta, Hyppoach rhamnoides, Filipendula ulmaria, Betula pendula, Polygonium odoratum, Brugmansi graveolens (ralf), Rhus toxicodenta, Armoraica ristica, Ficus benjaminii, Sluffera sp., Pelagonium zonale, Allium sp., Asimina triloba, Lippa dulcis, Epilobium augustifolium, Brugmansia suaveolens (old), Brugmansia suaveolens (young), Xanthosoma sagittifolium (leaf), Xanthosoma sagittifolium (stem), Monstera deliciosa, Aglaonema commutatus, Dieffenbachia leopoldii, Anthurium andreanum, Syngoniurn podophyllum, Dracaena fragrans, Ananas comosus, Strelitzia reglinae, Dieffenbachia segiunae, Syngoniurn aurutum, Dracaena sp., Hhaemanthus katharina, Anthurium altersianum, Spathiphyllum grandiflorum, Spathiphyllum cochlearispaturn, Monstera pertusa, Anthurium magnificurn, Anthurium hookeri, Anthurium elegans, Calathea zebrina, Yucca elephantipes, Bromelia balansae, Musa textilis (Leaf), Musa textilis (Stem), Myrthus communis, Olea olcaster, Olea europaea, Verium oleander, Cocculus laurifolius, Microsorium punctatum, Ficus sp., Senseviera sp., Adansonia digitata, Boechimeria boloba, Piper nigrum, Phymatosorus scolopendria, Tumera ulmifolia, Nicodemia diversifolia, Tapeinochilos spectabilis, Rauwolfia tetraphylla, Ficus elastica, Cycas cirinalis, Caryota ureus, Cynnamonum zeylonicum, Aechmea luddemoniana, Foenix zeulonica, Ficus benjamina, Ficus pumila, Murraya exotica, Trevesia sungaica, Clerodendrurn speciossicum, Actinidi colonicta, Paeonia lactiflora, Paeonia suffructicisa, Quercus imbricaria, Iris alida, Portulaca olleracea, Poligonum aviculare, Iris pseudocarpus, Allium nutans, Allium fistulosum, Antericum ramosum, Veratrum nigrum, Poligornun latifolia, Hosta lancefolia, Hosta zibalda, Echinops sphae, Paeonia daurica, Inula hilenium, Trambe pontica, Digitalis lutea, Bactisia australis, Austolachia australis, Hissopus zeraucharicus, Feucrium hamedris, Sedum album, Heraclelum pubescens, Origanurn vulgare, Cachris alpina, Haser trilobum, Matteucia strutiontoris, Sedum telchium, Bocconia cordata, Hiuga rentans, Talictrum minus, Anemona japonica, Clematis rectae, Talictrum sp., Alchemilla sp., Potentilla alba, Poterium sangiusorba, Minispermum dauricum, Oxobachus nictogenea, Armoracea rusticana, Cramble cardifolia, Agrimonia eupatora, Uschusa sp., Polymonium ceruleum, Valeriana officinalis, Pulmonaria molissima, Stachis lanata, Coronolla varia, Platicada grandiflora, Lavandula officinalis, Vincetocsicum officinalis, Acolypha hispida, Gnetum guemon, Psychotria nigropunctata, Psychotria metbacteriodomasica, Cobiaeum varilarturn, Phyllanthus grandifolium, Pterigota alata, Pachyra affinis, Sterulia elata, Phylidendron speciosus, Pithecelobium unguis, Sanchezia nobilis, Oreopanax capitata, Ficus triangularis, Pigelia pennata, Piper chaba, Laurus nobilis, Erythrinia caffra, Metrosideros excelsa, Osmanthus spp., Cupressus sempervirens, Jacobinia sp., Senecio platifilla, Livistona fragrans, Tetraclinis articulata hinensis, Eucaliptus rudis, Podocarpus spinulosus, Eriobotria japonica, Gingko biloba, Rhododendron spp., Thuja occidentalis, Fagopyrum suffruticosum, Geum macrophyllum, Magnolia cobus, Vinca minor, Convalaria majalis, Corylus avelana, Barbaric sp., Rosa multiflora, Ostrea carpinifolia, Ostrea connote, Quercus rubra, Tulip tree, Sorbus aucuparia, Betula nigra (leaf), Betula nigra (flower), Castanea sativa, Bergenia crassifolia, Artemisia dracunculus, Ruta graveolens, Quercus nigra, Schisandra chinensis, Betula alba, Sambucus niora, Gentiana cruciata, Encephalaris horridum, Phebodium aureum, Microlepia platphylla, Ceratoramia mexicana, Stepochlaena tenuifolia, Adianthum trapezieformis, Adianthum radiatum, Lycodium japonicum, Aessopteria crasifolia, Asplenium australasicum, Agatis robusta, Osmunda regalis, Osmundastrum claytonionum, Phyllitis scolopendrium, Polyschium braunii, Crytomium fortunei, Dryopteris filis-max, Equisetum variegatum, Anthyrium nopponicum, Anthyrium filis-femina, Parthenosicus tricuspidata, Ligustum vulgare, Charnaeciparis pisifera, Rosa cocanica, Citinis coggriaria, Pinus strobus, Celtis occidentalis, Picea schrenkiana, Cyclonia oblonga, Ulmus pumila, Euonomus verrucosa, Deutria scabra, Mespilus germanica, Quercus castanufolia, Euonomus europea, Seruginea suffruticisa, Keyleiteria paniculata, Sering a josiceae, Zelcova, carpinifolia, Abies cephalonica, Taccus bacata, Taxus cuspidata, Salis babilonics, Thuja occidentalis, Actinidia colomicta, Magonia agrifolia, Aralis mandshurica, Luglands nigra, Euonimus elata, Princepia sp., Forsitsia europea, Sorbocotoneaster sp., Morus alba, Crategus macrophyllum, Eucomia ulurifolia, Sorbus cominicta, Philodendron amurense, Comus mass, Korria japonica, Parrotia persica, Jasminum frutocarus, Sulda sanganea, Pentaphylloides fruticosa, Sibirea altaiensis, Cerasus japonica, Kolkwitzia amabilis, Amigdalus nana, Acer mandshurica, Salix tamarisifolia, Amelanchier spicata, Cerasus maghabab, Prunus cerasifera. Coryllus ayelana, Acer tataricum, Viburnum opulus, Siring a vulgaris, Fraxinus exelsior, Quercus trojana, Chaernomelis superba, Pinus salinifolia, Berberis vulgaris, Cotoneaster horisontalis, Cotoneaster fangianus, Fagus silvatica, Pinus pumila, Pinus silvestris and Berberis thungergi.

Another interesting group of plants that can be considered as plants and/or potential plants of the invention comprise the plants that are indigenous to arid regions, for example, those located between 35° north latitude and 35° south latitude. In accordance with the present invention potential extracts and extracts of the invention can be obtained from plants selected from the group comprising: the agave, Agavaceae, family including such members as: Yucca elata, Y. breviflora, Agave deserti, A. chrysantha, Dasylirion wheeleri; the buckwheat, Polygonaceae, family, such as Eriogonum fasciculatum; the crowfoot, Ranunculaceae, family, such as Delphinium scaposum, Anemone tuberosa and D. parishii; the poppy, Papaveraceae, family, including Platystemon californicus, Argemone pleiacantha, Corydalis aurea, Eschschoizia californica and Ar. corymbosa; members of the mustard, Cruciferae, family, such as Dithyrea californica, Streptanthus carinatus and Lesquerella gordoni; members of the legume, Leguminosae, family, such as Acacia greggii, Prosopis velutina, A. constrica, Senna covesii, Cercidium floridum, C. microphyllum, Lotus huminstratus, Krameria parvifolia, Parkinsonia aculeata, Calliendia eriophylla, Lupinus arizonicus, Olyneya tesota, Astragalus lentiginosus, Psorothamunus spinosus and Lupinus sparsiflorus; members of the loasa family, Loasaceae, including Mentzelia involucrata, M. pumila and Mohavea Confertiflora; members of the cactus, Cactaceae, family, such as Carnegiea gigantia, Opuntia leptocaulis, Ferocactus wislizenii, 0. bigelovii, 0. pheacantha, 0. versicolor, 0. fulgida, Echinocereus engelmannii, Mammillaria microcarpa, 0. basilaris, Stenocereins thurberi, 0. violacea, M. tetrancistra, 0. ramosissima, 0. acanthocarpa, E. pectinatins and 0. arbuscula; members of the evening primrose, Onagraceae, family, such as Oenothera deltoides, Camissonia claviformis and Oe. primiveris; members of the milkweed, Asclepiadaceae, family, including Asclepias erosa, A. sublata and Sarcostemma cynanchoides; members of the borage, Boraginaceae, family, such as Cryptantha augusti folia and Amsinckia intermedia; members of the sunflower, Compositae, family, including Baccharis sarothroides, Monoptiilon belloides, Erieron divergens, Zinnia acerosa, Melampodium leucanthan, Chaenactis fremontii, Calycoseris wrightii, Malacothrix californica, Helianthus annus, H. niveus, Geraea canescens, Hymenothrix wislizenii, Encelia farinosa Psilostrophe cooperi, Baileva multiradiata, Bebbia iuncea, Senecio douglasii, Trixis californica, Machaeranthera tephrodes, Xylorhiza tortifolia, Cirsiinm neomexicanum, Antennaria parviflora and Ch. douglasii; members of the caltrop, Zygophyllaceae, family, including Lama tridentata and Kallstroemia grandiflora; members of the mallow, Malvaceae, family, including Hibiscus coulteri, H. denudatus and Sphaeralcea ambigua; members of the phlox, Polemoniaceae, family, such as Luanthus aureus; members of the unicorn plant, Martyniaceae, family, such as Proboscidiea altheaefolia; members of the gourd, Cucurbitaceae, family, such as Cucurbita digitata; members of the lily, Lilaceae, family, including Calochortus kennedyi, Dichelostemma pulchellum, Allium macropetalum and Hesperocallis indulata; members of the ocotillo, Fouquieriaceae, family, including Fouquieria splendens; members of the figwort, Scrophulariaceae, family, such as Castilleja sp., Penstemon parryi and Orthocarpus purpurascens; members of the acanthus, Acanthaceae, family, including Anisacanthus thurberi, Justicia californica and Ruellia nudiflora; members of the four o'clock, Nyctaginaceae, family, such as Allionia incarnata, Abronia villosa and Mirabilis multiflora; members of the geranium, Geraniaceae, family, including Erodium cicutarium; members of the waterleaf, Hydrophyllaceae, family, such as Nama demissum, Phacelia bombycina and Ph. distans; members of the bignonia, Bignoniaceae, family, such as Chilopsis linearis; members of the vervain, Verbenaceae, family, including Glandularia gooddugii and Verbena neomexicana; members of the mint, Labiatae, family, such as Hyptis emoryi and Salvia columbariae; members of the broomrape, Orobanchaceae, family, such as Orobanche cooperi; members of the portulaca, Portulaceae, family, such as Talinum auriantiacum; members of the carpet-weed, Aizoaceae, family, such as Sesuvium verrucosum; members of the flax, Linaceae, family, such as Linum lewisii; members of the potato, Solanaceae, family, including Nicotiana trigonophylla and Physalis lobata; and members of the cochlospermum, Cochlospermaceae, family, such as Amoreuxia palmatifida.

Pre-Harvest Treatment

Once a potential plant is selected, a pre-harvest treatment is selected, wherein the treatment can be water or water in combination with a stressor, elicitor, or inducor. One skilled in the art would appreciate to perform the procedure with water and then with a series of stressors in order to determine whether the potential plant becomes an extract of the invention which demonstrates inhibitory activity against one or more extracellular proteases.

In one embodiment, this invention relates to altering the amount and/or composition of extracellular protease inhibitory activity by stressing a plant by chemical elicitors which act as stressor agent and activated defence plants pathways as mechanical wounding, drought, heat, or cold before tissue collection and extraction.

In one embodiment, stress involves exposing plants to a solution of one or more chemical elicitors to induce defense metabolic pathways and secondary metabolites prior to collection of plant tissues. Known chemical elicitors reported in the literature include ozone, hydrogen peroxide, jasmonic acid and its derivatives, arachidonic acid, salicylic acid and ester derivatives, alpha- and gamma-linoleic acids, volicitin, peptides, oligopeptides, saccharides, oligosaccharides such as chitosan, and synthetic chemicals such as Benzo-1,2,3-thiadiazole-7-carbathioic acid S-methyl ester (BTH).

A stressor may be one or more organic compounds. Some exemplary compounds that may be used as a stressor include Jasmonic acid, Jamonic acid lower alkyl esters, α-linoleic acid, α-linoleic acid lower alkyl esters, γ-linoleic acid, γ-linoleic acid lower alkyl esters, Arachidonic acid, Arachidonic acid lower alkyl esters, salicylic acid.

A stressor may be able to induce abiotic stresses in plants. Thus, for example, plants can be treated with one or more chemical or mechanical stresses prior to tissue collection. Mechanical stress can be performed twelve hours to ten days prior to tissue collection. In one embodiment, mechanical stress can be performed one day to three days prior to tissue collection. In one embodiment, mechanical stress can be performed three to six days prior to tissue collection. In one embodiment, mechanical stress can be performed four to eight days prior to tissue collection. In one embodiment, mechanical stress can be performed six to ten days prior to tissue collection.

Chemical stress can be induced by spraying plant material once or more than once with an aqueous or alcoholic solution of the chemical elicitor one hour to 10 days prior to tissue collection. In one embodiment, chemical stress can be induced one day to three days prior to harvesting the plant tissue; in one embodiment, chemical stress can be induced two to four days prior to harvesting the plant tissue; in one embodiment, chemical stress can be induced five to ten days prior to harvesting the plant tissue.

A chemical stress can be added by feeding a plant with an aqueous or alcoholic solution of the chemical. Likewise, the plants can be stressed by airborne transport of the chemical agents one hour to ten days prior tissue collection. In one embodiment, plants can be treated by spray one day before collection. In one embodiment, such chemical stress can be induced one hour to three days prior to harvesting the plant tissue; in one embodiment, such chemical stress can be induced two to eight days prior to harvesting the plant tissue; in one embodiment, such chemical stress can be induced five to ten days prior to harvesting the plant tissue.

Any combination of the above-mentioned stressors and treatment regiemes can be employed to induce the production or enhanced production of one or more extracellular proteases. One skilled in the art would be able to determine from the results of the assay against the panel of extracellular proteases whether it is desirable to follow one or more of the stressor regiemes.

Harvesting the Plant Material for Extraction and Optional Storage Treatment

The plant material may be used immediately after pre-harvest treatment, or it may be desirable to store the plant material for a period of time, prior to performing the extraction procedure(s). In one embodiment, the plant material could be treated prior to storage. In such cases, the treatment could include drying, freezing, lyophilisizing, or some combination thereof.

Following treatment to prepare the plant material for storage, the plant material may be stored for an extended period of time, prior to contacting the plant material with the first solvent. In one embodiment the plant material is stored less than one week. In one embodiment the plant material is stored from one week to one month. In one embodiment the plant material is stored from one month to six months. In one embodiment the plant material is stored from four months to one year. In one embodiment the plant material is stored longer than one year.

The Extraction Process

As depicted in FIG. 1, there are generally three basic extraction processes which can be performed in sequence to generate potential pre-extracts. The procedure for each Extraction process entails contacting the solid plant material with a solvent with adequate mixing and for an amount of time to ensure adequate exposure of the solid material to the solvent to enable inhibitory activity to be taken up by the solvent. Solvent A, B and C generally represent separate classes of solvents, for example, aqueous, alcoholic and organic. They are generally applied in a polar to non-polar order. They can be applied in a non-polar to polar order, however, in each case the solid matter must be dried prior to contacting the solid matter with the subsequent solvent. The liquid is then separated from the solid (insoluble) matter by a process known to those skilled in the art, to generate two fractions: the liquid fraction which is a potential pre-extract and a solid fraction.

The term “liquid” is used to denote a distinction from the solid, insoluble matter. Thus, a liquid, which may be converted to a gas or function in a gaseous form, as in the case with steam, for example can serve as a solvent. Likewise, other non-solid solvents may be used such as highly viscous liquids or other gaseous solvents, some of which can then be converted into a liquid phase.

A liquid solvent may also indicate a composition or a mixture of solvents. Common examples include a buffered aqueous solution, such as a TRIS-HCl buffer, or an ethanol/methanol combination.

In one embodiment, selected parts of a plant (which can be fresh, dried or frozen) can be crushed either mechanically, using a grinder or any device to break plant parts into small particles, or by freezing them in liquid nitrogen. In another embodiment, plant particles can be extracted with an aqueous TRIS-HCl buffer at pH 6-8, in one embodiment pH 7, from 30 minutes to 8 hours, in one embodiment 30 min to 2 hours, at a temperature between 4 to 50° C., in one embodiment 4 to 25° C.; in one embodiment, 4-10° C. In one embodiment, extraction can be performed at 4° C. for 30 minutes.

The solid material can be separated from the solvent by centrifugation, filtration or any other means known to those of skill in the art to separate solids from a solution, to yield aqueous, alcoholic or organic extract, a potential pre-extract. These potential pre-extracts can be tested directly by a panel of extracellular proteases for the ability to inhibit extracellular protease activity, and/or subjected to further separation procedures to generate a potential extract as described below.

The remaining solid can be contacted with a second solvent, such as an alcoholic solvent and a cosolvent, methanol or water In one embodiment, ethanol is used as alcoholic solvent, wherein the range of ethanol:methanol, ranges from 50:50 to 85:15, and 10 minute to one hour, in one embodiment 15 to 30 minute extraction time, at a temperature range of 4 to 25° C. in one embodiment, 4 to 10° C. in one embodiment, and 4° C. in another embodiment. Adequate contact of the solvent with the plant material can be encouraged by shaking the solid suspension for 15 min to 24 hour at a temperature ranging from 4 to 50° C.

The alcoholic extract is recovered and separated from the solids by centrifugation (the material which is insoluble in alcohol is used for organic extraction(s)). The potential pre-extract can be dried using a lyophilizer, a speed vac, a rotary evaporator, or a vacuum pump and dried under vacuum in order to remove the solvent. The dried extract can be dissolved in Tis-HCl buffer wherein the pH is between pH 6 to pH 8, in one embodiment and at pH 7 in one emodiment, and assayed against the panel of extracellular proteases for its bioactivity or, as in the case of the aqueous extract, the alcoholic extract can be treated to obtain purified extracts, as described below.

The organic extract can be obtained by shaking the residual solid for one to twenty-four hours in one embodiment, for one to fifteen hours in one embodiment, one to eight in one embodiment, one to four in one embodiment, with an organic solvent such as diethylether, hexane, dichloromethane, or ethylacetate. The solid can be separated by centrifugation or by filtration (regular or suction) and the organic solvent removed by distillation or by using a rotating evaporator. The organic extract can be dissolved in an aqueous buffer, or a mixture of an aqueous buffer and a suitable solvent (such as dimethylsulfoxide), to evaluate its bioactivity. In one embodiment the organic extracts are prepared using dichloromethane as the solvent of extraction, and the extraction is performed at room temperature for 2 hours.

Are included in the invention extracts prepared by all known large, medium and small-scale 30 methods to prepare extracts.

Determination of Extracellular Protease Inhibitory Activity in an Extract

In order to prepare various embodiments of the invention, (i.e., extracts, compositions and formulations with extracellular protease inhibitory activity) one requires techniques for measuring qualitatively and/or quantitatively the presence of such inhibitory activity. One skilled in the art would appreciate that there are numerous methods and techniques for measuring such activity, that can be used to determine, for example, which extracts are of interest and to follow the processing of the active ingredient(s) giving rise to such activity.

Currently, there are several assays to measure MMP, elastase andcathepsins activity (for a review of these methods, see Murphy and Crabbe, In Barrett (ed.) Methods in Enzymology. Proteolytic Enzymes: Aspa rtic Acid and Metallopeptidases (New York: Academic Press, 1995)-248: 470. One method, the gelatinolytic assay, is based on the degradation of radio-labelled type I collagen. Although this method is relatively sensitive, it requires the use of radio-labelled specific substrates.

Another widely-used technique is the zymography assay. In this assay, MMP, elastase and cathepsins activity is detected by the presence of negatively-stained bands following electrophoresis in substrate-impregnated SDS polyacrylamide gels. The zymography assay is a sensitive and quantitative method for the detection of various MMPs, elastase, cathepsins and TACE in biological samples; nonetheless, it is labour intensive and has a low dynamic range. Zymography, moreover, is not suitable to measure the intrinsic net activity in biological samples: SDS dissociates MMP-TIMP complexes and activates latent enzyme forms. This is particularly important since matrix degradation ultimately depends on the ratio of free active gelatinase to latent proenzyme or TIMP-complexed forms.

A microtitreplate assay has been developed recently (Pacmen et al., (1996) Biochem. Pharm. 52: 105-111). This assay provides measurement of net biological enzymatic activity of MMP, does not require a radioisotope safety environment, and could be used efficiently for routine measurement of inhibitory activity of MMP; however, it is not likely to be highly efficient as a diagnostic test since the incubation times are long and the sensitivity is much lower than that obtained by standard zymography and radio-labelled substrate assays.

Other methods used auto-quenched fluorogenic substrates. Many fluorogenic substrates have been designed for the quantification of MMPs, elastase, and cathepsins activity throught fluorescent level variation mesuring (reviewed by Nagase and Fields (1996) Biopolymers 40: 399-416).

Fluorescence polarization assays were based on the principle that when fluorescent molecules are excited with plane polarized light, they will emit light in the same polarized plane provided that the molecule remains stationary throughout the excited state. However, if the excited molecule rotates or tumbles during the excited state, then light is emitted in a plane different from the excitation plane. If vertically polarized light is used to excite the fluorophore, the emission light intensity can be monitored in both the original vertical plane and also the horizontal plane. The degree to which the emission intensity moves from the vertical to horizontal plane is related to the mobility of the fluorescently labeled molecule. If fluorescently labeled molecules are very large, they move very little during the excited state interval, and the emitted light remains highly polarized with respect to the excitation plane. If fluorescently labeled molecules are small, they rotate or tumble faster, and the resulting emitted light is depolarized relative to the excitation plane. Therefore, FP can be used to follow any biochemical reaction which results in a change in molecular size of a fluorescently labeled molecule (e.g. protein-DNA interactions; immunoassays; receptor-ligand interactions; degradation reactions). (Adapted from Bolger R, Checovich W. (1994) Biotechniques 17(3):585-9).

Another method uses the fluorescent activated substrate conversion (FASO) assay described in Canadian Patent No. 2,189,486 (1996) and in St-Pierre et al., (1996) Cytometry 25: 374-380.

The Commercial Process for Preparing Extracts of the Invention

Extracts of the invention can be prepared on a commercial scale by repeating the extraction process that results in an optimal composition of extracts demonstrating an inhibitory activity of interest. As demonstrated in FIG. 3, one would simply scale-up the procedure and include steps of quality control to ensure reproducible results for the resulting extracts.

Methods of Purifting or Fractionating Active Ingredients from Plant Extracts

There are a number of techniques well known in the art for isolating protease inhibitors from natural sources. For example, For example, purifications can be performed using centrifugation, ultracentrifugation, filtration, liquid or gas phase chromatography (including size exclusion, affinity, etc.) with or without high pressure, lyophylisation, evaporation, precipitation with various “carriers” (PVPP, carbon, antibody, etc.), or any combination thereof. One skilled in the art, would appreciate how to use the following options, in a sequential fashion, in order to enrich each successive fraction in the activity of interest by following its activity throughout the purification procedure, using one of the assays for the inhibitory activity against an extracellular protease of interest, as defined above.

The present invention also includes compounds, chemicals, active principles, and purified or concentrated extracts that could be obtained by purification, partial purification, and/or fractionation of plant extracts that are subject of the invention. Purification, partial purification, and/or fractionation can be achieved by any methods known by those skilled in the art. These methods include, but are not limited to: solid-liquid extraction, liquid-liquid extraction, solid-phase extraction (SPE), membrane and ultrafiltration, dialysis, chromatography, selective precipitation, electrophoresis, and solvent concentration.

Solid-liquid extraction means include the use of all possible solvents known from those in the art, and covers the use of supercritical solvents, soxhlet extractors, vortex shaker, ultrasounds and any other means to enhance extraction, as well as recovery by filtration, centrifugation and any related methods as described in the literature (R. J. P. Carmen, Natural Products Isolation, Humana Press, 1998). The solvent is selected from the group consisting of, but not limited to, hydrocarbon, chlorinated solvents, organic esters, organic ethers, alcohols, water, and mixtures thereof. In the case of supercritical fluid extraction, the invention also covers the use of modifiers as described in V. H. Bright, Eé Pé McNally, Supercritical Fluid Technology, ACS Symp. Ser. Vol. 488, ch. 22, 1999.

Liquid-liquid extraction means include the use of any mixture of solvents known from those in the art, including solvents under supercritical conditions. Typical solvents include, but are 30 not limited to, hydrocarbon, chlorinated solvents, organic esters, organic ethers, alcohols, water, and all possible aqueous solutions. The liquid-liquid extraction can be effected manually, semi-automated or completely automated, and the solvent can be removed or concentrated by any usual techniques known from those in the art (S. Ahuja. Handbook of Bioseparations, Academic Press, 2000).

Solid-phase extraction (SPE) means include techniques using cartridges, columns or any other devices used in this technique and known in the art. The sorbents that may be used with this method include but are not limited to silica gel (normal phase), reverse phase silica gel (modified silica gel), ion-exchange resins, and fluorisil. The invention also includes the use of scavenger resins or any others trapping reagents attached to solid supports derived from organic or inorganic macromolecular materials to remove selectively active ingredients or any constituents from said extracts.

Membrane, reverse osmosis and ultrafiltration means include the use of all types of membranes known from those in the art, as well as the use of pressure, vacuum, centrifugal force, and/or any other means that can be utilized in membrane and ultrafiltration processes (S. Ahuja, Handbook of Bioseparations, Academic Press, 2000).

Dialysis means include membranes having molecular weight cut-offs varying from less than 0.5 KDa to larger than 50 KDa. The invention also covers the recovery of purified and/or fractionated extracts from either the dialys ate or the retentate by any means known in the art including but not limited to evaporation, reduced pressure evaporation, distillation, vacuum distillation, and lyophilization.

Chromatographic means include all means of carrying out chromatography known by those skilled in the art and described in G. Sofer, L. Hagel, Handbook of Process Chromatography, Academic Press, 1997. Fractionation, partial purification, and/or purification can be carried out by but not limited to regular column chromatography, flash chromatography, high performance liquid chromatography (HPLC), medium pressure liquid chromatography (MPLC), supercritical fluid chromatography (SFC), countercurrent chromatography (CCC), moving bed chromatography, simulated moving bed chromatography, expanded bed chromatography, and planar chromatography. With every chromatographic methods, sorbents that may be used include but is not limited to silica gel, alumina, fluorisil, cellulose and modified celluloses, all possible modified silica gels, all types of ion-exchange resins, all types of size exclusion gels and any other sorbents known from those skilled in the art and described in T. Hanai. HPLC: A Practical Guide. RSC Press, UK 1999. The present invention also includes the use of two or more solvent gradients to effect the fractionation, partial purification, and/or purification of said active extracts in any chromatographic methods. The solvents that may be utilized include but are not limited to hexanes, pentane, petroleum ethers, cyclohexane, heptane, diethyl ether, methanol, ethanol, isopropanol, propanol, butanol, isobutanol, tert-butanol, water, dichloromethane, dichloroethane, ethyl acetate, tetrahydrofurane, dioxane, tert-butyl methyl ether, acetone, and 2-butanone. When water or and aqueous phase is used, it may contains certain amounts of iorganic or organic salts and the pH may be adjusted to different values with an acid or a base to enhance fractionation and/or purification.

In the case of planar chromatography, the present invention includes the use of all variants of this type of chromatography including but not limited to one- and two dimension thin-layer chromatography (1D- and 2D-TLC), high performance thin-layer chromatography (HPTLC), and centrifugal thin-layer chromatography (centrifugal TLC).

In the case of countercurrent chromatography (CCC), the present invention includes the use of manual, semi-automated, and automated systems, and the use of all possible solvents and solvent combinations necessary to effect fractionation and/or purification of said active extracts as described in W. D. Conway, R. J. Petroski, Modem Countercurrent Chromatography, ACS Symp. Ser. Vol. 593, 1995. Solvent removal and/or concentration can be effected by any means known by those skilled in the art, including but not limited to reduced pressure evaporation, evaporation, reduced pressure distillation, distillation, and lyophilization.

The present invention includes the fractionation, partial purification, and purification of said active plant extracts by expanded bed chromatography, moving and simulated moving bed chromatography, and any other related methods known by those skilled in the art and described in G. Sofer, L. Hagel, Handbook of Process Chromatography, Academic Press, 1997 and S. Ahuja, Handbook of Bioseparations, Academic Press, 2000.

Selective precipitation means includes the use of all possible solvents and solvent combinations, the use of temperature changes, the addition of precipitent and/or modifiers, and/or modifying the pH by adding a base or an acid to effect a selective preceipation of active principles or any other constituents.

Further, the present invention covers the fractionation, partial purification, and purification of said active plant extracts by electrophoresis and other related techniques known tothose skilled in the art.

The invention also includes the fractionation, partial purification, and/or purification of said active plant extracts by steam distillation, hydrodistillation, or any other related methods of distillation known from those in the art (L. M. Harwood, C. J. Moody, Experimental Organic Chemistry, Blackwell Scientific Publications, UK, 1989).

The process of purifying the active component(s) also includes the concentration of purified or partially purified chemicals, active ingredients, active principles by solvent removal of said plant extracts and/or fractionated plant extracts, and/or purified plant extracts. The techniques of solvent removal are known to those skilled in the art and include but are not limited to rotary evaporation, distillation (normal and reduced pressure), centrifugal vacuum evaporation (speed-vac), and lyophilization.

One embodiment of the invention includes the concentration of chemicals, active ingredients, active principles by solvent removal of said plant extracts and/or fractionated plant extracts, and/or purified plant extracts. The techniques of solvent removal are known to those skilled in the art and include but are not limited to rotary evaporation, distillation (normal and reduced pressure), centrifugal vacuum evaporation (speed-vac), and lyophilization.

To get a better understanding of the invention described herein, the following examples are set forth. It should be understood that these examples are for illustrative purposes only. Therefore, they should not limit the scope of this invention in any way.

EXAMPLES Example I Preparation of Stressed and Non-stressed Plant Extracts

Pre-Harvest TreatmentAerian parts of a living plant are sprayed with an aqueous solution of gamma linolenic acid (6,9,12-Octadecatrienoic acid, Sigma L-2378) (stress G) or arachidonic acid (5,8,11,14-Eicosatetraenoic acid, Sigma A-3925) (stress A) (400 μM in water with 0.125% (v/v) Triton X-100) to completely cover the leaves.

Harvest Solid SI and Optional Storage Treatment

Twenty to twenty-four hours after the stress, more than 4 grams of leaves, stems, fruit, flowers, seeds or other plant parts are harvested and frozen immediately in dry ice, then transferred as soon as possible to a −20° C. freezer until use. Plant materials may be stored at −20° C. for a long period of time, more than a year, without losing inhibitory activity. Temperature is monitored to ensure a constant condition.

Stressed and non-stressed plant specimens are collected as wet samples and stored at −20° C. for various periods of time, and are submitted to a process which generats 3 subtractions: aqueous, ethanolic and organic fractions. Complete extraction process are performed in a continuous cycle using the following steps. An initial 5g of plant specimen is homogenized in liquid nitrogen with a blender. The resulting powder is weighed.

Extraction Process I: Aqueous Extraction

To each 4.5 grams of plant powder, 12 ml of a cold solution of 100 mM Tris, pH 7.0 is added. The mixture is thoroughly vortexed for 2 minutes. The mixture is kept on ice for 30 minutes and vortexed after each 10 minute period of time. The sample is centrifuged in a Corex™ 30 ml tube for 5 minutes at 4500 rpm. The resulting supernatant is decanted in a 15 ml tube after filtration with a Miracloth™ filter. This extract is therefore referred as the Potential Pre-Extract A. The pellet, referred as Solid S2, is kept for ethanolic extraction.

The aqueous extract (Potential Pre-Extract A) is further purified in order to determine its extracellular protease inhibition capability. The Potential Pre-Extract A is purified by size-exclusion chromatography, wherein the aqueous extract is chromatographed on a calibrated Sephadex G-25 column (1×10 cm) using a 20 mM Tris-HCl, 150 mM NaCl, pH 7.5 buffer as eluant. Fractions corresponding to compounds that seem to have molecular weight (MW) less than 1500 daltons (D) are pooled to constitute the purified aqueous extract that is tested for inhibitory activity in an assay as described in Example II.

Prior to this analysis, the extract is treated with 10% gelatin-Sepharose (Pharmacia Biotech, Uppsala, Sw.) in order to remove unspecific enzyme ligands. To 1 mL of extract, 100 μL of gelatin-Sepharose resin is added in a microassay tube, the solution in the tube is mixed, kept on ice for 30 minutes, and then centrifuged 5 minutes at 5,000 rpm. The supernatant is removed and used directly for assays.

Extraction Process II: Alcholic Extraction

To the pellet, Solid S2, collected from the previous aqueous extraction, 12 ml of cold ethanol:methanol (85:15) is added and the mixture is thoroughly vortexed for 2 minutes. The mixture is kept on ice for 30 minutes and vortexed every 10 minutes. The sample is centrifuged in a Corex™ 30 ml tube for 5 minutes at 4,500 rpm. The resulting supernatant is decanted in a 15 ml tube after filtration with a Miracloth™ filter. The pellet, referred as Solid S3 is kept for the subsequent organic extraction. This extract is therefore referred as the Potential Pre-Extract B.

The ethanolic extract, Potential Pre-Extract B, is purified by liquid/liquid extraction prior to analysis by enzymatic assay. For this purpose, 1 ml of ethanolic extract is evaporated under vacuum, dissolved in 150 μA of dimethylsulfoxide (DMSO), and completed to a final volume of 1.5 ml with Tris buffer (final concentration: Tris-HC120 mM; pH 7.5). Four ml of hexane is added to the Tris phase in a glass tube and the tube is thoroughly vortexed, then allowed to form a biphasic liquid. The organic phase is removed and the extract is submitted to a second round of liquid/liquid extraction. The aqueous phase is removed and treated with 10% gelatin-Sepharose (Pharmacia Biotech, Uppsala, Sw.) to remove unspecific enzyme ligands prior to conducting subsequent assays. To 1 ml of extract, 100 μl of gelatin-Sepharose resin is added in a microassay tube, the tube is mixed, kept on ice for 30 minutes, and then centrifuged 5 minutes at 5,000 rpm. Supernatant is removed and used directly for assays as described in Example II.

Extraction Process III: Organic Extraction

To the pellet, Solid S3, collected from previous ethanolic extraction, 12 ml of cold dichloromethane is added and the mixture is thoroughly vortexed for 2 minutes. The mixture is kept on ice for 30 minutes and vortexed after each 10 minutes period. The sample is centrifuged in a Corex™ 30 ml tube for 5 minutes at 4,500 rpm. The resulting supernatant is decanted in a 15 ml glass tube after filtration with a Miracloth™ filter. The final pellet is discarded. The organic solvent is evaporated under vacuum and the phase is dissolved with dimethylsulfoxide (DMSO). This extract is therefore referred as the Potential Pre-Extract C, which was further purified by solid phase extraction prior to analysis by enzymatic assay.

In order to assay the Potential Pre-Extract C, the organic extract is diluted 1:10 in a solution of DMSO:Methanol:Tris (20 mM, pH 7.5) (10:50:40) (Solution A), ie, 220 of extract is added to 2.0 ml of solution A. After 10 seconds of vigorous vortex, the mix is sonicated for 10 seconds. Dissolved extracts are subsequently applied to a solid phase extraction plate (Discovery SPE-96, Sigma Chemical Co, St-Louis, Mo.). After initial conditioning of the columns with 1 ml of methanol, columns are equilibrated with solution A, and extract samples are deposited on the columns. Elution is completed with solution A (final volume of 2 ml) and this fraction is used directly in assays as described in Example II.

Example II In vitro Enzyme Inhibition Assays

The inhibitory activity of sample compositions towards human MMP-1, human MMP-2, human MMP-3, human MMP-9, human cathepsin-B, human cathepsin-D, human cathepsin-G, human cathepsin-L, human cathepsin-K, human leukocyte elastase (HLE), bacteria clostripain and bacteria subtilisin can be determined using either fluorogenic substrates or the FASC assay.

Measurement of Human MMP-1, -2, -3 and -9 Activity with Fluorogenic Peptidic Substrates

MMP-1, -2, -9 are purified from natural sources (human immortalized cell lines: 8505C (Deutsche Sammlung von Mikroorganismen and Zellkulturen GmbH) for MMP-1, HT-1080 (ATCC, Manassas, Va.) for MMP-2 and THP-1 (ATCC, Manassas, Va.) for MMP-9) as described in literature and based on protocols found in I. M. Clark: <<Matrix metalloproteinases protocol>>, Humana Press (2001). Recombinant human MMP-3 is overexpressed in E. Coli and purified according to Windsor L J, Steele D L (2001), Methods Mol Biol 151:191-205. Proteolytic activity of these proteases is evaluated with the assay based on the cleavage of auto-quenched peptide substrate: (MCA-Pro-Leu-Gly-Leu-Dpa-Ala-Arg-NH2.TFA [Dpa ═N-3-(2,4-dinitrophenyl)-L-2,3-diaminopropionyl]) for MMP-1, -2, and -9; and, MCA-Arg-Pro-Lys-Pro-Val-Glu-Nva-Trp-Arg-Lys(DNP)—NH2 (DNP=2,4-dinitrophenyl; Nva=L-norvaline) for MMP-3 (Calbiochem, San Diego, Calif.). In the intact peptide, Dpa or DNP quenches the MCA fluorescence. Cleavage of the peptide causes release of the fluorescent MCA group which is then quantitated on a fluorometer (Gemini XS, Molecular Devices, Sunnyvale, Calif.). The assay is performed in TNCZ assay buffer (20 mM Tris-HCl; NaCl 150 mM; CaCl2 5 mM; ZnCl2 0.5 mM; pH 7.5) with human purified proteases (I. M. Clark: <<Matrix metalloproteinases protocol>>, Humana Press (2001). The substrate, primarily dissolved in DMSO is then redissolved in TNCZ buffer for the assay. In a typical assay, 10 μl of purified enzyme (1-50 ng) and 5 μl of dissolved substrate (final concentration of 10 μM) is mixed in a final volume of 75 μl (completed with TNCZ). All assays were performed in 96 well plate and the reaction is started by the addition of substrate. Assays are measured (excitation 325 nm, emission 392 nm) for 20, 40 and 60 minutes.

Measurement of Human Cathepsin L and K Activity with Fluorogenic Peptidic Substrate.

Human recombinant cathepsins L and K are overexpressed in P. Pastoris according to Krupa J C, Mort J S. (2000), Anal Biochem 283(1):99-103. The assay is similar to the previous except for the auto-quenched peptidic substrate: Z-Arg-Phe-AMC, 2HCl (Bachem California, Torrance, Calif.) and reaction buffer. Assays for Cathepsin L are performed in 20 mM acetate pH 5.5, 1 mM EDTA buffer and assays for Cathepsin K in 20 mM acetate pH 4.2, 1 mM EDTA. Assays are monitored with fluorometer settled at excitation 380 nm/emission 460 nm wavelengths (Krupa J C, Mort J S. (2000), Anal Biochem 283(1):99-103).

Measurement of Human MMP-9, Cathepsin B, Cathepsin G, and Human Leukocyte Elastase (HLE) Activity Using the FASC Assay

Human Cathepsin B and G and human leukocyte elastase are obtained from Calbiochem (San Diego, Calif.). Human MMP-9 is purified as previously described. The assay is based on the method described in Canadian Patent No. 2,189,486 (1996) and in St-Pierre et al., (1996) Cytometry 25:374-380. For the assay, 5 μl of the purified enzyme (1-100 ng), 5 μl of concentrated buffer solution (20 mM Tris-HCl; NaCl 150 mM; CaCl2 5 mM; ZnCl2 0.5 mM; pH 7.5), and 5 μl of gelatin-FITC beads are typically used in a final volume of 100 μl. The assay is performed by incubation of the reaction mixture for 90 minutes at 37° C. The reaction is stopped by the transfer of the mix in 0.5 ml of 20 mM Tris, 150 mM NaCl; pH 9.5 buffer. This tube is analyzed in a flow cytometer (Epics MCL, Beckman Coulter, Mississauga, Ontario) as described in Canadian Patent No. 2,189,486 (1996).

Measurement of Human Cathepsin D, Cathepsin B, Cathepsin G and HLE Activity with a Fluorogenic Proteic Substrate

Cathepsin D is purified from human MCF-7 cells according to Stewart A J, Piggott N H, May F E, Westley B R. (1994), Int J Cancer 57(5):715-8. Cathepsin B, Cathepsin G and HLE are obtained as previously described. The activities of Cathepsin D, Cathepsin B, Cathepsin G and HLE are measured by an assay based on the increase of fluorescence of a proteic substrate (Haemoglobin in the case of Cathepsin D and B and beta-casein in the case of Cathepsin G and HLE) heavily labelled with Alexa-488 dye (Molecular Probes, Eugene, Or). The substrate, when highly labelled with the dye, will almost quench the dye fluorescence. Cleavage of the substrate will result in an increase of the fluorescence which can be measured with a spectrofluorometer, and which is proportional to protease activity. Typically, 10 μA of purified human Cathepsin D, Cathepsin B, Cathepsin G or HLE (10-50 ng) and 10 μl of Hemoglobin-Alexa488 or beta-casein-Alexa488 (100 ng) are assayed in final volume of 75 μA adjusted with 20 mM citrate pH 3.3 buffer in the case of Cathepsins D and B or TNCZ buffer in the case of Cathepsin G and HLE. The reaction is performed as already described except that the fluorescence is read at excitation 488 nm/emission 525 nm wavelengths.

Subtilisin Assay

Subtilisin (isolated from B. Subtilisis) is purchased from Fluka. Assays are performed with a fluorogenic peptide (Z-Gly-Gly-Leu-AMC, Bachem California, Torrance, Calif.) as already described for MMPs with the following modification: the assay is buffered with 20 mM Tris, 150 mM NaCl; pH 7.5 and the results are read at excitation 380 nm/emission 460 nm wavelengths.

Clostripain Assay

Clostripain from Clostridium histolyticum (Worthington Lakewood, N.J.) is prepared and activated as described by manufacturer's protocol. The activity is determined by using Z-Arg-Arg-AMC, 2HCl (Calbiochem, San Diego, Calif.) as a fluorogenic peptidic substrate and the incubation buffer is 75 mM phosphate, pH 7.6. The reaction is performed as already described except that the fluorescence is read at excitation 380 nm/emission 460 nm wavelengths.

Extract Inhibition Assay

Before a typical assay, aqueous extracts prepared as described in Example I are preincubated with 1:10 of gelatin-Sepharose 4B™ for 30 minutes to remove fluorescence quenching. For the ethanolic extract, an initial hexane extraction is performed and samples are treated with 1:10 of gelatin-Sepharose 4B™ to remove quenching.

In a typical fluorescent assay, 10 μl of purified enzyme at concentrations previously mentioned for the enzymatic assay, 5 μl of dissolved fluorogenic peptide or 10 μl of dissolved fluorescent proteic substrate (final concentration of 10 μM) and 40 μl., of the aqueous, ethanolic or organic extract to be tested and prepared as described in Example I are mixed in a final volume of 75 μl (completed with TNCZ for fluorogenic peptide substrate assay or 20 mM citrate pH 3.3 buffer for fluorescent protein substrate assay). All assays are performed in 96 well plate and the reaction is started by the addition of substrate. Assays are measured (excitation 325 nm, emission 392 nm for peptide and excitation 488 nm/emission 525 nm wavelengths for protein) for 20, 40 and 60 minutes. Activity and inhibition values are determined from the increase in fluorescence.

For the FASC assay, 35 μl of the treated extract prepared as described in Exampla 1, 5 μl of the purified enzyme prepared as described previously, 5 μl of concentrated buffer solution (TNCZ), and 5 of gelatin-FITC beads are typically used. The initial step of the assay is the incubation of the reaction without beads for a 30 minutes period on ice to allow the binding of inhibitors to enzyme. Fluorescent beads are added and the reaction mix is incubated for 90 minutes at 37° C. The reaction is stopped by transfer of the mix in 0.5 ml of 20 mM Tris, 150 mM NaCl; pH 9.5 buffer. This tube isanalyzed in the flow cytometer (Epics MCL, Beckman Coulter, Mississauga, Ontario) as described in Canadian Patent Application No. 2,189,486 (1996).

Results of the inhibition studies are shown in Tables 1-12. Table 1 reports the inhibition of human MMP-1 by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. Table 2 reports the inhibition of human MMP-2 by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. Table 3 reports the inhibition of human MMP-3 by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. Table 4 reports the inhibition of human MMP-9 by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. Table 5 reports the inhibition of human Cathepsin B by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. Table 6 reports the inhibition of human Cathepsin D by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. Table 7 reports the inhibition of human Cathepsin G by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. Table 8 reports the inhibition of human Cathepsin L by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. Table 9 reports the inhibition of human Cathepsin K by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. Table 10 reports the inhibition of HLE by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. Table 12 reports the inhibition of bacteria subtilisin by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. Table 11 reports the inhibition of bacterial clostripain by aqueous (A), ethanolic (R) and organic (S) extracts for exemplary stressed (A and G) and non-stressed (T) plant sources. The inhibition is reported as percentage (%) of inhibition of substrate degradation as compared with the degradation without extract.

Example III Examplary Purification of Inhibitory Activityfound in an Extract

Extracts were separated by HPLC on an Agilent 1100 system (San Fernando, Calif.). Briefly, 100 μl of a crude extract prepared as described in Example I was applied on a C18 reverse-phase column (Purospher RP-18 5 μm, 4.0×125 mm (HP), Agilent, San Fernando, Calif.). Elution of compounds was achieved with a linear gradient of 10-85% acetonitrile. Fractions were collected, evaporated, resuspended in aqueous buffer and then reanalysed for their inhibition activity on specific enzymes as already described. Fractions of interest (demonstrating a biological activity) where then reisolated at a larger scale for further analysis and characterization.

The invention being thus described, it will be obvious that the same may be varied in many ways. Such variations are not to be regarded as a departure from the spirit and scope of the invention, and all such modifications as would be obvious to one skilled in the art are intended to be included within the scope of the following claims.

Claims

1-7. (canceled)

8. An extracellular protease-inhibiting plant extract, said plant extract capable of inhibiting the activity of one or more matrix metalloproteinase (MMP), one or more cathepsin, human leukocyte elastase (HLE), or a combination thereof, and said plant extract derived from Aconitum napellus L.; Acorus calamus L.; Agrostis alba; Alchemilla mollis L.; Allium cepa L.; Allium porrum L.; Allium sativum L.; Allium schoenoporasum; Allium tuberosum; Aloe vera; Althaea officinalis L.; Amaranthus caudathus L.; Ambrosia artemisiifolia L.; Anethum graveolens L.; Angelica sinensis; Anthemis tinctoria L.; Arctostaphylos uva-ursi; Aronia melanocarpa (Michx) Ell.; Aronia×prunifolia; Artemisia dracunculus L.; Asarum europaeum; Avena sativa L.; Begonia mannii; Begonia polygonoides; Beta vulgaris L.; Beta vulgaris L. subsp. Maritima; Borago officinalis L.; Brassica napus L.; Brassica oleracea L.; Brassica rapa L.; Bromus inermis; Capsicum annuum L. var. annuum; Chaerophyllum bulbosum L.; Chenopodium quinoa Willd.; Cichorium endivia L.; Circium arvense L. Scop.; Citrullus lanatus; Cornus canadensis L.; Daucus carota; Dioscorea batatas; Dirca palustris L.; Dolichos lablab L.; Fagopyrum esculentum Moench; Fagopyrum tartaricum; Foeniculum vulgare Mill.; Fragaria×ananassa; Frangula alnus Mill.; Gentiana lutea L.; Geranium×cantabrigiense; Glycine max; Glycyrrhiza glabra L.; Hamamelis virginiana L.; Helianthus strumosus; Heliotropium arborescens L.; Hordeum vulgare L. subsp. vulgare; Humulus lupulus L.; Hypomyces lactiflorum; Iberis sempervirens; Juniperis communis L.; Kochia scoparia L.; Lactuca sativa L.; Lentinus edodes; Lotus comiculatus L.; Lunaria annua L.; Manihot esculenta; Matricaria recutita L.; Melilotus albus Medik.; Melilotis officinalis; Mentha×piperita; Oenothera biennis L.; Origanum majorana L.; Origanum vulgare L.; Panax quinquefolius L.; Pastinaca sativa L. subsp. sativa; Petroselinum crispum; Phalaris canariensis L.; Phaseolus vulgaris L. subsp. vulgaris; Physostegia virginiana L. Benth.; Phytolacca decandra; Pimpinella anisum L.; Pisum sativum L.; Plantago major L.; Polygonum persicaria L.; Potentilla anserina L.; Poterium sanguisorba L.; Pyrus communis L.; Raphanus raphanistrum L.; Rheum rhabarbarum; Ribes nigrum L.; Ribes rubrum L.; Rodgersia spp.; Rosmarinus officinalis L.; Rubus allegheniensis; Rubus idaeus L.; Rubus occidentalis L.; Rubus thibetanus; Rumex crispus L.; Rumex scutatus L.; Ruta graveolens L.; Salvia officinalis L.; Sambucus canadensis L.; Sambucus ebulus L.; Saponaria officinalis L.; Satureja montana L.; Setaria italica; Silene vulgaris; Solanum melongena L.; Solidago canadensis; Solidago virgaureae; Solidago×hybrida; Sorghum caffrorum; Sorghum dochna; Stellaria media (L.) Vill.; Stephanandra incise; Tamarindus indica L.; Tanacetum cinerariifolium; Tanacetum vulgare L.; Taraxacum officinale; Teucrium chamaedrys L.; Thymus citriodorus; Thymus fragrantissimus; Thymus praecox subsp arcticus; Thymus pseudoanuginosus; Thymus vulgaris; Trifolium incarnatum L.; X Triticosecale spp.; Tropaeolum majus L.; Tsuga canadensis L.; Tsuga diversifolia; Vaccinium angustifolium Aiton.; Vigna unguiculata; Vinca minor; Vitis sp.; Zea mays L. subsp. mays or Zingiber officinale.

9. The extracellular protease-inhibiting plant extract according to claim 8, wherein said plant extract is an Avena sativa L.; Capsicum annuum L. var. annuum; Chenopodium quinoa Willd.; Geranium×cantabrigiense; Glycine max; Helianthus strumosus; Iberis sempervirens; Juniperis communis L.; Lotus corniculatus L.; Melilotus albus Medik.; Pastinaca sativa L. subsp. sativa; Potentilla anserina L.; Rheum rhabarbarum; Rubus idaeus L.; Salvia officinalis L.; Sambucus canadensis L.; Saponaria officinalis L.; Solidago canadensis; Solidago virgaureae; Taraxacum officinale; Thymus citriodorus; Thymus vulgaris; X Triticosecale spp.; Tropaeolum majus L.; Tsuga canadensis L.; Vaccinium angustifolium Aiton; Zea mays L. subsp. mays or Zingiber officinale extract.

10. The extracellular protease-inhibiting plant extract according to claim 8, wherein said plant extract is capable of inhibiting the activity of one or more MMP and is an Aconitum napellus L.; Acorus calamus L.; Agrostis alba; Alchemilla mollis L.; Allium cepa L.; Allium porrum L.; Allium sativum L.; Allium schoenoporasum; Allium tuberosum; Aloe vera; Althaea officinalis L.; Amaranthus caudathus L.; Ambrosia artemisiifolia L.; Anethum graveolens L.; Anthemis tinctoria L.; Arctostaphylos uva-ursi; Aronia melanocarpa (Michx) Ell.; Aronia×prunifolia; Artemisia dracunculus L.; Avena sativa L.; Beta vulgaris L.; Beta vulgaris L. subsp. Maritima; Borago officinalis L.; Brassica napus L.; Brassica oleracea L.; Brassica rapa L.; Bromus inermis; Capsicum annuum L. var. annuum; Chaerophyllum bulbosum L.; Chenopodium quinoa Willd.; Cichorium endivia L.; Circium arvense L. Scop.; Citrullus lanatus; Cornus canadensis L.; Daucus carota; Dioscorea batatas; Dirca palustris L.; Dolichos lablab L.; Fagopyrum esculentum Moench; Fagopyrum tartaricum; Foeniculum vulgare Mill.; Fragaria×ananassa; Gentiana lutea L.; Glycine max; Glycyrrhiza glabra L.; Hamamelis virginiana L.; Helianthus strumosus; Heliotropium arborescens L.; Hordeum vulgare L. subsp. vulgare; Humulus lupulus L.; Hypomyces lactiflorum; Juniperis communis L.; Kochia scoparia L.; Lactuca sativa L.; Lentinus edodes; Lotus comiculatus L.; Lunaria annua L.; Manihot esculenta; Matricaria recutita L.; Melilotus albus Medik.; Melilotis officinalis; Mentha X piperita; Oenothera biennis L.; Origanum majorana L.; Origanum vulgare L.; Panax quinquefolius L.; Pastinaca sativa L. subsp. sativa; Petroselinum crispum; Phalaris canariensis L.; Phaseolus vulgaris L. subsp. vulgaris; Phytolacca decandra; Pimpinella anisum L.; Pisum sativum L.; Plantago major L.; Polygonum persicaria L.; Potentilla anserina L.; Poterium sanguisorba L.; Pyrus communis L.; Raphanus raphanistrum L.; Rheum rhabarbarum; Ribes nigrum L.; Rosmarinus officinalis L.; Rubus allegheniensis; Rubus idaeus L.; Rubus occidentalis L.; Rubus thibetanus; Rumex crispus L.; Rumex scutatus L.; Ruta graveolens L.; Salvia officinalis L.; Sambucus canadensis L.; Sambucus ebulus L.; Saponaria officinalis L.; Satureja montana L.; Setaria italica; Silene vulgaris; Solanum melongena L.; Solidago canadensis; Solidago virgaureae; Sorghum caffrorum; Sorghum dochna; Stellaria media (L.) Vill.; Tanacetum cinerariifolium; Tanacetum vulgare L.; Taraxacum officinale; Teucrium chamaedrys L.; Thymus citriodorus; Thymus fragrantissimus; Thymus praecox subsp arcticus; Thymus pseudoanuginosus; Thymus vulgaris; Trifolium incarnatum L.; X Triticosecale spp.; Tropaeolum majus L.; Tsuga canadensis L.; Tsuga diversifolia; Vaccinium angustifolium Aiton.; Vigna unguiculata; Vinca minor; Vitis sp.; Zea mays L. subsp. mays or Zingiber officinale extract.

11. The extracellular protease-inhibiting plant extract according to claim 8, wherein said plant extract is capable of inhibiting at least MMP-9 activity and is an Aconitum napellus L.; Acorus calamus L.; Agrostis alba; Alchemilla mollis L.; Allium cepa L.; Allium porrum L.; Allium sativum L.; Allium schoenoporasum; Allium tuberosum; Althaea officinalis L.; Amaranthus caudathus L.; Ambrosia artemisiifolia L.; Anthemis tinctoria L.; Arctostaphylos uva-ursi; Aronia melanocarpa (Michx) Ell.; Aronia×prunifola; Artemisia dracunculus L.; Avena sativa L.; Beta vulgaris L.; Beta vulgaris L. subsp. Maritima; Borago officinalis L.; Brassica napus L.; Brassica oleracea L.; Brassica rapa L.; Bromus inermis; Capsicum annuum L. var. annuum; Chaerophyllum bulbosum L.; Chenopodium quinoa Willd.; Cichorium endivia L.; Citrullus lanatus; Cornus canadensis L.; Daucus carota; Dioscorea batatas; Dirca palustris L.; Dolichos lablab L.; Fagopyrum esculentum Moench; Fagopyrum tartaricum; Foeniculum vulgare Mill.; Fragaria×ananassa; Gentiana lutea L.; Glycine max; Glycyrrhiza glabra L.; Hamamelis virginiana L.; Helianthus strumosus; Heliotropium arborescens L.; Hordeum vulgare L.; Humulus lupulus L.; Hypomyces lactiflorum; Juniperis communis L.; Lactuca sativa L.; Lentinus edodes; Lotus comiculatus L.; Lunaria annua L.; Manihot esculenta; Matricaria recutita L.; Melilotus albus Medik.; Meliotis officinalis; Mentha X piperita; Oenothera biennis L.; Origanum majorana L.; Origanum vulgare L.; Panax quinquefolius L.; Pastinaca sativa L. subsp. sativa; Phalaris canariensis L.; Phaseolus vulgaris L.; Phytolacca decandra; Pimpinella anisum L.; Pisum sativum L.; Plantago major L.; Polygonum persicaria L.; Potentilla anserina L.; Poterium sanguisorba L.; Pyrus communis L.; Raphanus raphanistrum L.; Rheum rhabarbarum; Ribes nigrum L.; Rosmarinus officinalis L.; Rubus idaeus L.; Rubus allegheniensis; Rubus occidentalis L.; Rumex crispus L.; Rumex scutatus L.; Ruta graveolens L.; Salvia officinalis L.; Sambucus canadensis L.; Sambucus ebulus L.; Satureja montana L.; Setaria italica; Solanum melongena L.; Solidago canadensis; Solidago virgaureae; Sorghum caffrorum; Sorghum dochna; Tanacetum cinerariifolium; Tanacetum vulgare L.; Taraxacum officinale; Teucrium chamaedrys L.; Thymus citriodorus; Thymus fragrantissimus; Thymus pseudoanuginosus; Thymus vulgaris; Trifolium incarnatum L.; X Triticosecale spp.; Tropaeolum majus L.; Tsuga canadensis L.; Tsuga diversifolia; Vaccinium angustifolium Aiton.; Vigna unguiculata; Vinca minor; Vitis sp.; Zea mays L. subsp. mays or Zingiber officinale extract.

12. The extracellular protease-inhibiting plant extract according to claim 8, wherein said plant extract is capable of inhibiting at least MMP-9 activity and is an Avena sativa L.; Capsicum annuum L. var. annuum; Chenopodium quinoa Willd.; Glycine max; Helianthus strumosus; Juniperis communis L.; Lotus comiculatus L.; Melilotus albus Medik.; Pastinaca sativa L. subsp. sativa; Potentilla anserina L.; Rheum rhabarbarum; Rubus idaeus L.; Salvia officinalis L.; Sambucus canadensis L.; Solidago canadensis; Solidago virgaureae; Taraxacum officinale; Thymus citriodorus; Thymus vulgaris; X Triticosecale spp.; Tropaeolum majus L.; Tsuga canadensis L.; Vaccinium angustifolium Aiton; Zea mays L. subsp. mays or Zingiber officinale extract.

13. The extracellular protease-inhibiting plant extract according to claim 8, wherein said plant extract is capable of inhibiting at least cathepsin B activity and is an Aconitum napellus L.; Alchemilla mollis L.; Allium cepa L.; Allium sativum L.; Allium schoenoporasum; Allium tuberosum; Althaea officinalis L.; Angelica sinensis; Anthemis tinctoria L.; Arctostaphylos uva-ursi; Aronia melanocarpa (Michx) Ell.; Aronia×prunifolia; Artemisia dracunculus L.; Asarum europaeum; Begonia mannii; Begonia polygonoides; Beta vulgaris L.; Beta vulgaris L. subsp. Maritima; Borago officinalis L.; Brassica napus L.; Brassica oleracea L.; Brassica rapa L.; Capsicum annuum L. var. annuum; Citrullus lanatus; Daucus carota; Dirca palustris L.; Fragaria×ananassa; Glycine max; Glycyrrhiza glabra L.; Hamamelis virginiana L.; Heliotropium arborescens L.; Humulus lupulus L.; Hypomyces lactiflorum; Iberis sempervirens; Kochia scoparia L.; Lentinus edodes; Lunaria annua L.; Origanum majorana L.; Origanum vulgare L.; Panax quinquefolius L.; Pastinaca sativa L. subsp. sativa; Petroselinum crispum; Phaseolus vulgaris L.; Physostegia virginiana L. Benth.; Phytolacca decandra; Plantago major L.; Ribes nigrum L.; Rubus allegheniensis; Rubus idaeus L.; Rubus occidentalis L.; Rumex crispus L.; Salvia officinalis L.; Sambucus canadensis L.; Saponaria officinalis L.; Satureja montana L.; Silene vulgaris; Solidago canadensis; Solidago×hybrida; Sorghum caffrorum; Tamarindus indica L.; Tanacetum vulgare L.; Taraxacum officinale; Thymus praecox subsp arcticus; Thymus vulgaris; Tropaeolum majus L.; Tsuga diversifolia; Vaccinium angustifolium Aiton.; Vitis sp. or Zea mays L. subsp. mays extract.

14. The extracellular protease-inhibiting plant extract according to claim 8, wherein said plant extract is capable of inhibiting at least cathepsin B activity and is a Capsicum annuum L. var. annuum; Glycine max; Iberis sempervirens; Pastinaca sativa L. subsp. sativa; Rubus idaeus L.; Salvia officinalis L.; Sambucus canadensis L.; Saponaria officinalis L.; Solidago canadensis; Taraxacum officinale; Thymus vulgaris; Tropaeolum majus L.; Vaccinium angustifolium Aiton. or Zea mays L. subsp. mays extract.

15. The extracellular protease-inhibiting plant extract according to claim 8, wherein said plant extract is capable of inhibiting at least HLE activity and is an Aconitum napellus L.; Acorus calamus L.; Alchemilla mollis L.; Allium sativum L.; Allium tuberosum; Aloe vera; Althaea officinalis L.; Amaranthus caudathus L.; Anethum graveolens L.; Anthemis tinctoria L.; Arctostaphylos uva-ursi; Aronia melanocarpa (Michx) Ell.; Aronia×prunifolia; Artemisia dracunculus L.; Avena sativa L.; Begonia mannii; Begonia polygonoides; Beta vulgaris L.; Beta vulgaris L. subsp. Maritima; Borago officinalis L.; Brassica napus L.; Brassica oleracea L.; Brassica rapa L.; Bromus inermis; Capsicum annuum L. var. annuum; Chaerophyllum bulbosum L.; Chenopodium quinoa Willd.; Cichorium endivia L.; Circium arvense L. Scop.; Citrullus lanatus; Cornus canadensis L.; Daucus carota; Dirca palustris L.; Dolichos lablab L.; Fagopyrum esculentum Moench; Fagopyrum tartaricum; Foeniculum vulgare Mill.; Fragaria×ananassa; Frangula alnus Mill.; Gentiana lutea L.; Geranium×cantabrigiense; Glycine max; Glycyrrhiza glabra L.; Hamamelis virginiana L.; Heliotropium arborescens L.; Hordeum vulgare L.; Humulus lupulus L.; Hypomyces lactiflorum; Iberis sempervirens; Juniperis communis L.; Lactuca sativa L.; Lentinus edodes; Matricaria recutita L.; Mentha X piperita; Oenothera biennis L.; Origanum majorana L.; Origanum vulgare L.; Pastinaca sativa L. subsp. sativa; Petroselinum crispum; Phalaris canariensis L.; Phaseolus vulgaris L.; Phytolacca decandra; Pimpinella anisum L.; Plantago major L.; Polygonum persicaria L.; Poterium sanguisorba L.; Ribes nigrum L.; Ribes rubrum L.; Rodgersia spp.; Rosmarinus officinalis L.; Rubus idaeus L.; Rubus occidentalis L.; Rubus thibetanus; Rumex crispus L.; Rumex scutatus L.; Ruta graveolens L.; Salvia officinalis L.; Sambucus canadensis L.; Sambucus ebulus L.; Satureja montana L.; Solanum melongena L.; Sorghum caffrorum; Sorghum dochna; Stellaria media (L.) Vill.; Stephanandra incise; Tanacetum cinerariifolium; Tanacetum vulgare L.; Taraxacum officinale; Teucrium chamaedrys L.; Thymus citriodorus; Thymus fragrantissimus; Thymus praecox subsp arcticus; Thymus pseudoanuginosus; Thymus vulgaris; X Triticosecale spp.; Tropaeolum majus L.; Tsuga diversifolia; Vaccinium angustifolium Aiton.; Vitis sp. or Zea mays L. subsp. mays extract.

16. The extracellular protease-inhibiting plant extract according to claim 8, wherein said plant extract is capable of inhibiting at least HLE activity and is an Avena sativa L.; Capsicum annuum L. var. annuum; Chenopodium quinoa Willd.; Geranium×cantabrigiense; Glycine max; Iberis sempervirens; Juniperis communis L.; Pastinaca sativa L. subsp. sativa; Rubus idaeus L.; Salvia officinalis L.; Sambucus canadensis L.; Taraxacum officinale; Thymus citriodorus; Thymus vulgaris; X Triticosecale spp.; Tropaeolum majus L.; Vaccinium angustifolium Aiton. or Zea mays L. subsp. mays extract.

17. The extracellular protease-inhibiting plant extract according to claim 8, wherein said plant extract is derived from a stressed plant.

18. The extracellular protease-inhibiting plant extract according to claim 8, wherein said plant extract is selected from a library of extracellular protease-inhibiting plant extracts, said library prepared by a process comprising the steps of:

(a) selecting a group of plants;
(b) harvesting plant material from each plant in said group of plants;
(c) obtaining an extract from said plant material by contacting the plant material with an aqueous, an ethanolic or an organic solvent, or a combination thereof, thereby providing a plurality of plant extracts;
(d) analysing each of said plant extracts for the presence of inhibitory activity against one or more MMP, one or more cathepsin, HLE, or a combination thereof, and
(e) selecting plant extracts having inhibitory activity to provide said library of extracellular protease inhibiting plant extracts.

19. The extracellular protease-inhibiting plant extract according to claim 11, wherein said inhibitory activity is represented by a decrease of at least 20% in the activity of the one or more MMP, one or more cathepsin, HLE, or a combination thereof, in the presence of the plant extract compared to in the absence of the plant extract.

20. The extracellular protease-inhibiting plant extract according to claim 11, wherein said process further comprises stressing said group of plants prior to harvesting said plant material.

21. An extracellular protease-inhibiting composition comprising one or more plant extracts according to claim 8.

22. The extracellular protease-inhibiting composition according to claim 14, wherein each of said one or more plant extracts is derived from Avena sativa L.; Capsicum annuum L. var. annuum; Chenopodium quinoa Willd.; Geranium×cantabrigiense; Glycine max; Helianthus strumosus; Iberis sempervirens; Juniperis communis L.; Lotus corniculatus L.; Melilotus albus Medik.; Pastinaca sativa L. subsp. sativa; Potentilla anserina L.; Rheum rhabarbarum; Rubus idaeus L.; Salvia officinalis L.; Sambucus canadensis L.; Saponaria officinalis L.; Solidago canadensis; Solidago virgaureae; Taraxacum officinale; Thymus citriodorus; Thymus vulgaris; X Triticosecale spp.; Tropaeolum majus L.; Tsuga canadensis L.; Vaccinium angustifolium Aiton; Zea mays L. subsp. mays or Zingiber officinale.

23. A method of inhibiting an extracellular protease comprising contacting said extracellular protease with a plant extract according to claim 8.

24. The method according to claim 16, wherein said plant extract is an Avena sativa L.; Capsicum annuum L. var. annuum; Chenopodium quinoa Willd.; Geranium×cantabrigiense; Glycine max; Helianthus strumosus; Iberis sempervirens; Juniperis communis L.; Lotus comiculatus L.; Melilotus albus Medik.; Pastinaca sativa L. subsp. sativa; Potentilla anserina L.; Rheum rhabarbarum; Rubus idaeus L.; Salvia officinalis L.; Sambucus canadensis L.; Saponaria officinalis L.; Solidago canadensis; Solidago virgaureae; Taraxacum officinale; Thymus citriodorus; Thymus vulgaris; X Triticosecale spp.; Tropaeolum majus L.; Tsuga canadensis L.; Vaccinium angustifolium Aiton; Zea mays L. subsp. mays or Zingiber officinale extract.

25. The method according to claim 16, wherein said extracellular protease is a MMP and said plant extract is an Aconitum napellus L.; Acorus calamus L.; Agrostis alba; Alchemilla mollis L.; Allium cepa L.; Allium porrum L.; Allium sativum L.; Allium schoenoporasum; Allium tuberosum; Aloe vera; Althaea officinalis L.; Amaranthus caudathus L.; Ambrosia artemisiifolia L.; Anethum graveolens L.; Anthemis tinctoria L.; Arctostaphylos uva-ursi; Aronia melanocarpa (Michx) Ell.; Aronia×prunifolia; Artemisia dracunculus L.; Avena sativa L.; Beta vulgaris L.; Beta vulgaris L. subsp. Maritima; Borago officinalis L.; Brassica napus L.; Brassica oleracea L.; Brassica rapa L.; Bromus inermis; Capsicum annuum L. var. annuum; Chaerophyllum bulbosum L.; Chenopodium quinoa Willd.; Cichorium endivia L.; Circium arvense L. Scop.; Citrullus lanatus; Cornus canadensis L.; Daucus carota; Dioscorea batatas; Dirca palustris L.; Dolichos lablab L.; Fagopyrum esculentum Moench; Fagopyrum tartaricum; Foeniculum vulgare Mill.; Fragaria×ananassa; Gentiana lutea L.; Glycine max; Glycyrrhiza glabra L.; Hamamelis virginiana L.; Helianthus strumosus; Heliotropium arborescens L.; Hordeum vulgare L. subsp. vulgare; Humulus lupulus L.; Hypomyces lactiflorum; Juniperis communis L.; Kochia scoparia L.; Lactuca sativa L.; Lentinus edodes; Lotus comiculatus L.; Lunaria annua L.; Manihot esculenta; Matricaria recutita L.; Melilotus albus Medik.; Melilotis officinalis; Mentha X piperita; Oenothera biennis L.; Origanum majorana L.; Origanum vulgare L.; Panax quinquefolius L.; Pastinaca sativa L. subsp. sativa; Petroselinum crispum; Phalaris canariensis L.; Phaseolus vulgaris L. subsp. vulgaris; Phytolacca decandra; Pimpinella anisum L.; Pisum sativum L.; Plantago major L.; Polygonum persicaria L.; Potentilla anserina L.; Poterium sanguisorba L.; Pyrus communis L.; Raphanus raphanistrum L.; Rheum rhabarbarum; Ribes nigrum L.; Rosmarinus officinalis L.; Rubus allegheniensis; Rubus idaeus L.; Rubus occidentalis L.; Rubus thibetanus; Rumex crispus L.; Rumex scutatus L.; Ruta graveolens L.; Salvia officinalis L.; Sambucus canadensis L.; Sambucus ebulus L.; Saponaria officinalis L.; Satureja montana L.; Setaria italica; Silene vulgaris; Solanum melongena L.; Solidago canadensis; Solidago virgaureae; Sorghum caffrorum; Sorghum dochna; Stellaria media (L.) Vill.; Tanacetum cinerariifolium; Tanacetum vulgare L.; Taraxacum officinale; Teucrium chamaedrys L.; Thymus citriodorus; Thymus fragrantissimus; Thymus praecox subsp arcticus; Thymus pseudoanuginosus; Thymus vulgaris; Trifolium incarnatum L.; X Triticosecale spp.; Tropaeolum majus L.; Tsuga canadensis L.; Tsuga diversifolia; Vaccinium angustifolium Aiton.; Vigna unguiculata; Vinca minor; Vitis sp.; Zea mays L. subsp. mays or Zingiber officinale extract.

26. The method according to claim 16, wherein said extracellular protease is MMP-9 and said plant extract is an Aconitum napellus L.; Acorus calamus L.; Agrostis alba; Alchemilla mollis L.; Allium cepa L.; Allium porrum L.; Allium sativum L.; Allium schoenoporasum; Allium tuberosum; Althaea officinalis L.; Amaranthus caudathus L.; Ambrosia artemisiifolia L.; Anthemis tinctoria L.; Arctostaphylos uva-ursi; Aronia melanocarpa (Michx) Ell.; Aronia×prunifola; Artemisia dracunculus L.; Avena sativa L.; Beta vulgaris L.; Beta vulgaris L. subsp. Maritima; Borago officinalis L.; Brassica napus L.; Brassica oleracea L.; Brassica rapa L.; Bromus inermis; Capsicum annuum L. var. annuum; Chaerophyllum bulbosum L.; Chenopodium quinoa Willd.; Cichorium endivia L.; Citrullus lanatus; Cornus canadensis L.; Daucus carota; Dioscorea batatas; Dirca palustris L.; Dolichos lablab L.; Fagopyrum esculentum Moench; Fagopyrum tartaricum; Foeniculum vulgare Mill.; Fragaria×ananassa; Gentiana lutea L.; Glycine max; Glycyrrhiza glabra L.; Hamamelis virginiana L.; Helianthus strumosus; Heliotropium arborescens L.; Hordeum vulgare L.; Humulus lupulus L.; Hypomyces lactiflorum; Juniperis communis L.; Lactuca sativa L.; Lentinus edodes; Lotus corniculatus L.; Lunaria annua L.; Manihot esculenta; Matricaria recutita L.; Melilotus albus Medik.; Meliotis officinalis; Mentha X piperita; Oenothera biennis L.; Origanum majorana L.; Origanum vulgare L.; Panax quinquefolius L.; Pastinaca sativa L. subsp. sativa; Phalaris canariensis L.; Phaseolus vulgaris L.; Phytolacca decandra; Pimpinella anisum L.; Pisum sativum L.; Plantago major L.; Polygonum persicaria L.; Potentilla anserina L.; Poterium sanguisorba L.; Pyrus communis L.; Raphanus raphanistrum L.; Rheum rhabarbarum; Ribes nigrum L.; Rosmarinus officinalis L.; Rubus idaeus L.; Rubus allegheniensis; Rubus occidentalis L.; Rumex crispus L.; Rumex scutatus L.; Ruta graveolens L.; Salvia officinalis L.; Sambucus canadensis L.; Sambucus ebulus L.; Satureja montana L.; Setaria italica; Solanum melongena L.; Solidago canadensis; Solidago virgaureae; Sorghum caffrorum; Sorghum dochna; Tanacetum cinerariifolium; Tanacetum vulgare L.; Taraxacum officinale; Teucrium chamaedrys L.; Thymus citriodorus; Thymus fragrantissimus; Thymus pseudoanuginosus; Thymus vulgaris; Trifolium incarnatum L.; X Triticosecale spp.; Tropaeolum majus L.; Tsuga canadensis L.; Tsuga diversifolia; Vaccinium angustifolium Aiton.; Vigna unguiculata; Vinca minor; Vitis sp.; Zea mays L. subsp. mays or Zingiber officinale extract.

27. The method according to claim 16, wherein said extracellular protease is MMP-9 and said plant extract is an Avena sativa L.; Capsicum annuum L. var. annuum; Chenopodium quinoa Willd.; Glycine max; Helianthus strumosus; Juniperis communis L.; Lotus comiculatus L.; Melilotus albus Medik.; Pastinaca sativa L. subsp. sativa; Potentilla anserina L.; Rheum rhabarbarum; Rubus idaeus L.; Salvia officinalis L.; Sambucus canadensis L.; Solidago canadensis; Solidago virgaureae; Taraxacum officinale; Thymus citriodorus; Thymus vulgaris; X Triticosecale spp.; Tropaeolum majus L.; Tsuga canadensis L.; Vaccinium angustifolium Aiton; Zea mays L. subsp. mays or Zingiber officinale extract.

28. The method according to claim 16, wherein said extracellular protease is cathepsin B and said plant extract is an Aconitum napellus L.; Alchemilla mollis L.; Allium cepa L.; Allium sativum L.; Allium schoenoporasum; Allium tuberosum; Althaea officinalis L.; Angelica sinensis; Anthemis tinctoria L.; Arctostaphylos uva-ursi; Aronia melanocarpa (Michx) Ell.; Aronia×prunifolia; Artemisia dracunculus L.; Asarum europaeum; Begonia mannii; Begonia polygonoides; Beta vulgaris L.; Beta vulgaris L. subsp. Maritima; Borago officinalis L.; Brassica napus L.; Brassica oleracea L.; Brassica rapa L.; Capsicum annuum L. var. annuum; Citrullus lanatus; Daucus carota; Dirca palustris L.; Fragaria×ananassa; Glycine max; Glycyrrhiza glabra L.; Hamamelis virginiana L.; Heliotropium arborescens L.; Humulus lupulus L.; Hypomyces lactiflorum; Iberis sempervirens; Kochia scoparia L.; Lentinus edodes; Lunaria annua L.; Origanum majorana L.; Origanum vulgare L.; Panax quinquefolius L.; Pastinaca sativa L. subsp. sativa; Petroselinum crispum; Phaseolus vulgaris L.; Physostegia virginiana L. Benth.; Phytolacca decandra; Plantago major L.; Ribes nigrum L.; Rubus allegheniensis; Rubus idaeus L.; Rubus occidentalis L.; Rumex crispus L.; Salvia officinalis L.; Sambucus canadensis L.; Saponaria officinalis L.; Satureja montana L.; Silene vulgaris; Solidago canadensis; Solidago×hybrida; Sorghum caffrorum; Tamarindus indica L.; Tanacetum vulgare L.; Taraxacum officinale; Thymus praecox subsp arcticus; Thymus vulgaris; Tropaeolum majus L.; Tsuga diversifolia; Vaccinium angustifolium Aiton.; Vitis sp. or Zea mays L. subsp. mays extract.

29. The method according to claim 16, wherein said extracellular protease is cathepsin B and said plant extract is a Capsicum annuum L. var. annuum; Glycine max; Iberis sempervirens; Pastinaca sativa L. subsp. sativa; Rubus idaeus L.; Salvia officinalis L.; Sambucus canadensis L.; Saponaria officinalis L.; Solidago canadensis; Taraxacum officinale; Thymus vulgaris; Tropaeolum majus L.; Vaccinium angustifolium Aiton. or Zea mays L. subsp. mays extract.

30. The method according to claim 16, wherein said extracellular protease is HLE and said plant extract is an Aconitum napellus L.; Acorus calamus L.; Alchemilla mollis L.; Allium sativum L.; Allium tuberosum; Aloe vera; Althaea officinalis L.; Amaranthus caudathus L.; Anethum graveolens L.; Anthemis tinctoria L.; Arctostaphylos uva-ursi; Aronia melanocarpa (Michx) Ell.; Aronia×prunifolia; Artemisia dracunculus L.; Avena sativa L.; Begonia mannii; Begonia polygonoides; Beta vulgaris L.; Beta vulgaris L. subsp. Maritima; Borago officinalis L.; Brassica napus L.; Brassica oleracea L.; Brassica rapa L.; Bromus inermis; Capsicum annuum L. var. annuum; Chaerophyllum bulbosum L.; Chenopodium quinoa Willd.; Cichorium endivia L.; Circium arvense L. Scop.; Citrullus lanatus; Cornus canadensis L.; Daucus carota; Dirca palustris L.; Dolichos lablab L.; Fagopyrum esculentum Moench; Fagopyrum tartaricum; Foeniculum vulgare Mill.; Fragaria×ananassa; Frangula alnus Mill.; Gentiana lutea L.; Geranium×cantabrigiense; Glycine max; Glycyrrhiza glabra L.; Hamamelis virginiana L.; Heliotropium arborescens L.; Hordeum vulgare L.; Humulus lupulus L.; Hypomyces lactiflorum; Iberis sempervirens; Juniperis communis L.; Lactuca sativa L.; Lentinus edodes; Matricaria recutita L.; Mentha X piperita; Oenothera biennis L.; Origanum majorana L.; Origanum vulgare L.; Pastinaca sativa L. subsp. sativa; Petroselinum crispum; Phalaris canariensis L.; Phaseolus vulgaris L.; Phytolacca decandra; Pimpinella anisum L.; Plantago major L.; Polygonum persicaria L.; Poterium sanguisorba L.; Ribes nigrum L.; Ribes rubrum L.; Rodgersia spp.; Rosmarinus officinalis L.; Rubus idaeus L.; Rubus occidentalis L.; Rubus thibetanus; Rumex crispus L.; Rumex scutatus L.; Ruta graveolens L.; Salvia officinalis L.; Sambucus canadensis L.; Sambucus ebulus L.; Satureja montana L.; Solanum melongena L.; Sorghum caffrorum; Sorghum dochna; Stellaria media (L.) Vill.; Stephanandra incise; Tanacetum cinerariifolium; Tanacetum vulgare L.; Taraxacum officinale; Teucrium chamaedrys L.; Thymus citriodorus; Thymus fragrantissimus; Thymus praecox subsp arcticus; Thymus pseudoanuginosus; Thymus vulgaris; X Triticosecale spp.; Tropaeolum majus L.; Tsuga diversifolia; Vaccinium angustifolium Aiton.; Vitis sp. or Zea mays L. subsp. mays extract.

31. The method according to claim 16, wherein said extracellular protease is HLE and said plant extract is an Avena sativa L.; Capsicum annuum L. var. annuum; Chenopodium quinoa Willd.; Geranium×cantabrigiense; Glycine max; Iberis sempervirens; Juniperis communis L.; Pastinaca sativa L. subsp. sativa; Rubus idaeus L.; Salvia officinalis L.; Sambucus canadensis L.; Taraxacum officinale; Thymus citriodorus; Thymus vulgaris; X Triticosecale spp.; Tropaeolum majus L.; Vaccinium angustifolium Aiton. or Zea mays L. subsp. mays extract.

Patent History
Publication number: 20110217753
Type: Application
Filed: Aug 30, 2010
Publication Date: Sep 8, 2011
Applicant: BIOPHARMACOPAE DESIGN INTERNATIONAL INC. (Sainte-Foy)
Inventor: Benoit CYR (St. Augustin de Desmaures)
Application Number: 12/871,097
Classifications
Current U.S. Class: Enzyme Inactivation By Chemical Treatment (435/184); Organic Reactant (252/182.12)
International Classification: C12N 9/99 (20060101);