<<

USOO5885829A United States Patent (19) 11 Patent Number: 5,885,829 Mooney et al. (45) Date of Patent: Mar 23, 1999

54) ORAL TISSUES Hubbell, “Biomaterials in tissue engineering.” Bio/Technol ogy, 13:565-576, 1995. 75 Inventors: David J. Mooney; Robert B. Langer and Vacanti, "TiSSue engineering,” Science, Rutherford, both of Ann Arbor, Mich. 260:920–926, 1993. 73 Assignee: The Regents of the University of Laurencin et al., “Use of polyphosphaZenes for skeletal tissue regeneration,” J. Biomed. Mater. Res., 27:963-973, Michigan, Ann Arbor, Mich. 1993. Lesot et al., “Experimental Induction of odontoblast Differ 21 Appl. No.: 864,494 entiation and Stimulation During Reparative Processes.” 22 Filed: May 28, 1997 Cells and Materials, 3:201–217, 1993. Mikos et al., “Preparation of poly (glycolic acid) bonded Related U.S. Application Data Structures for cell attachment and transplantion, J. Biomed. Mat. Res., 27:183, 1993. 60 Provisional application No. 60/018.450, May 28, 1996. Mikos et al., “Prevascularization of porous biodegradable 51 Int. Cl...... C12N 5700; C12N 5/02; polymers,” Biotech. bioeng, 42:716–723, 1993. C12N 5/08; C12N 15/09 Mooney and Vacanti, "Tissue engineering using cells and 52 U.S. Cl...... 435/325; 424/49; 424/422; synthetic polymers,” Trans. Rey, 7:153–162, 1993. 424/435; 435/69.5; 435/374; 435/378 Mooney et al., “Design and fabrication of biodegradable 58 Field of Search ...... 435/69.1, 325, polymer devices to engineer tubular tissues, Cell Trans 435/69.4, 69.5, 69.6, 365, 393, 366, 374, plantation, 3:203, 1994. 378; 422/422, 423, 424, 435, 49, 85.1, Mooney et al., “Transplantation of hepatocytes using 93.7; 514/12, 21 porous, biodegradable Sponges,” Transplan. Proc., 56) References Cited 26(6):4025-4026, 1994. Mooney et al., “Biodegradable Sponges for Hepatocyte U.S. PATENT DOCUMENTS Transplantation,” J. Biomed. Mat. Res.,29:959-965, 1995. 4.963,489 10/1990 Naughton et al...... 435/1.1 Mooney et al., “Fabricating Tubular Devices From Polymers 5,032,508 7/1991 Naughton et al...... 435/32 of Lactic and Glycolic Acid for TiSSue Engineering,” Tiss. 5,266,480 11/1993 Naughton et al...... 435/371 Eng., 1:107–118, 1995. 5,443,950 8/1995 Naughton et al...... 435/1.1 5,518.915 5/1996 Naughton et al. ... 424/422 Mooney et al., “Stabilized Polyglycolic Acid Fibre-Based 5,567,612 10/1996 Vacanti et al...... 435/366 Tubes for Tissue Engineering,” Biomaterials, 17:115-124, 5,624,840 4/1997 Naughton et al. ... 435/395 1996. 5,759,830 6/1998 Vacanti et al...... 435/18O Mooney et al., “Engineering dental pulp-like tissue in 5,770,193 6/1998 Vacanti et al...... 424/93.7 vitro,” Biotechnol. Prog. 12(6):865–868, 1996. 5,770,417 6/1998 Vacanti et al...... 435/180 Mooney, et al., “Localized delivery of epidermal growth FOREIGN PATENT DOCUMENTS factor improves the Survival of transplanted hepatocytes,” Biotechnology and Bioengineering, 50(4):422-429, 1996. 40 40 872 7/1992 Germany. WO 88/03785 6/1988 WIPO. Mooney et al., “Novel approach to fabricate porous Sponges WO 92,07573 5/1992 WIPO. of poly(D.Llactic-co-glycolic acid) without the use of WO 92/16181 10/1992 WIPO. organic solvents.” Biomaterials, 17(14): 1417–1422, 1996. WO 98/22041 5/1998 WIPO. Mooney, et. al., “Tissue engineering: Tubular tissues,” In: OTHER PUBLICATIONS Yearbook of Cell and Tissue Transplantion, Lanza and Chick (Ed.), pp. 275-282, 1996. Bohl, K. et al., “Synthetic Extracellular Matrices for Engi neering Dental Pulp,' Abstract distributed a Fifth World Nakashima, “Induction of Dentin Formation on Canine Biomaterals Congress, Toronto, Canada, May 29, 1996. Amputated Pulp by Recombinant Human Bone Morphoge Gombotz and Pettit, “Biodegradable polymers for protein netic Protieins (BMP)-2 and -4,” J. Dent Res., and peptide drug delivery,” Bioconjugate Chem., 73:1515-1522, 1994. 6:332-351, 1995. Goshima et al., “The origin of bone formed in composite (List continued on next page.) grafts of porous calcium phosphate ceramic loaded with Primary Examiner Nancy Degen marrow cells,” Clin. Orthopod. Rel. Res., 269:274–283, 1991. Attorney, Agent, or Firm Arnold, White & Durkee Green et al., “Growth of cultured human epidermal cells into 57 ABSTRACT multiple epithelia suitable for grafting.” Proc. Natl. Acad. Sci. USA, 76:5665-5668, 1979. Disclosed are methods for regenerating dental and oral Gu, et al., “Expression of genes for bone morphogenetic tissues from viable cells using eX Vivo culture on a structural proteins and receptors in human dental pulp', Archives of matrix. The regenerated oral tissues and tissue-matrix prepa Oral Biology, 41(10):919–23, 1996. rations thus provided have both clinical applications in Hansbrough et al., “Evaluation of a biodegradable matrix dentistry and oral medicine and are also useful in in Vitro containing cultured human fibroblasts as a dermal replace toxicity and biocompatibility testing. ment beneath meshed skin grafts on athymic mice, Surgery, 4:438-446, 1992. 109 Claims, 11 Drawing Sheets 5,885,829 Page 2

OTHER PUBLICATIONS Yannas et al., “Synthesis and characterization of a model of extracellular matrix that induces partial regeneration of adult Putnam, and Mooney, "Tisue engineering using Synthetic mammalian skin,” Proc. Natl. Acad. Sci. USA, 86:933-937, extracellular matrices,” Nature Medicine 207):824-826, 1989. 1996. Rutherford et al., “Induction of Reparative Dentine Forma Bouvier, Joffre and Magloire, “In Vitro Mineralization of a tion in Monkeys by Recombinant Human Osteogenic Pro Three-Dimensional Collagen Matrix by Human Dental pulp tein-1, Archs Oral Biol, 38:571-576, 1993. Cells in the Presence of Chondroitin Sulphate.” Archs Oral Rutherford et al., “Platelet derived and insulin-like growth Biol, 35(4):301–309, 1990 factorS Stimulate regeneration of periodontal attachment in Rutherford et al., “Synergistic Effects of Dexamethasone on monkeys,” J. Perio Res., 27:285-290, 1992. Platelet-Derived Growth Factor Mitogenesis. In Vitro,” Rutherford et al., “The Time-Course of the Induction of Archs Oral Biol, 37(2):139–145, 1992. Reparative Dentine Formation in Monkeys by Recombinant Sasaki et al., “Inhibition of Wound Contraction bt Papaver Human Osteogenic Protein-1, Arch Oral Biol., ine: In Vitro Analysis with a Submucosal Tissue Model.” 39:833-838, 1994. Annals of Plastic Surgery, 27(6),Dec. 1991. Rutherford et al., “Transdentinal Stimulation of Reparative Suwa et al., “Inductive Effect of Bovine Bone Morphoge Dentine formation by Osteogenic Protein-1, Archs. Oral netic Protein on Human Dental Pulp Tissue. In Vitro,” Biol., 40:681–683, 1995. 13-Mammalian Biochem., 122:707, 1995, Abstract No. Rutherford et al., “Platelet-derived growth factor and dex 77691 W. amethasone combined with a collagen matrix induced regen Ueda, Ebata, and Kaneda, “In Vitro Fabrication of Bioarti eration of the periodontium in monkeys,” J. Clin. Perio, ficial Mucosa for Reconstruction of Oral Mucosa: Basic 20:537-544, 1993. Research and Clinical Application,” Annals of Plastic Sur Yannas et al., “Wound tissue can utilize a polymeric tem gery, 27(6), Dec. 1991. plate to Synthesize and functional extension of Skin,” Sci International Search Report dated Sep. 29, 1997 ence, 215:174-176, 1981. (UMIC:019P). U.S. Patent Mar 23, 1999 Sheet 1 of 11 5,885,829

U.S. Patent 5,885,829

U.S. Patent Mar 23, 1999 Sheet 3 of 11 5,885,829

s U.S. Patent Mar 23, 1999 Sheet 4 of 11 5,885,829 0.4

0.3 E. co E s 0.2 a s 0.1

0.0 O 10 20 30 40 50 60 70 Time (days) FIG. 4A 108 s if 107 Sl s C CO a 106 s CD

105 O 10 20 30 40 50 60 70 Time (days) FIG. 4B U.S. Patent Mar 23, 1999 Sheet S of 11 5,885,829

100

s 80 E s C 60 & d S3 40

f 20 85/15PLGA O O 10 20 30 40 50 60 Time (weeks) FIG. 5A

an S S S & s E

H YseSS .9 O S - Cl2 Vs Old 5 as s E s 8 O da

Time (days) FIG. 5B U.S. Patent Mar 23, 1999 Sheet 6 of 11 5,885,829

O 10 20 Time (min) FIG. 6 U.S. Patent Mar 23, 1999 Sheet 7 of 11 5,885,829

100 60 SeC 30 SeC 4.68 OOO 20 SeC 2 O

10 20 Time (minutes) FIG. 7A 100 COntrol 8 O Pre-Wet

6 O

4 O

2 O

5 10 15 Time (minutes) FIG. 7B U.S. Patent Mar 23, 1999 Sheet 8 of 11 5,885,829

100

80

60

40

20

O O 2 4 6 8 10 12 Time (wk) FIG. 8 80 4060

2 O

O 10 20 30 40 50 Time (days) FIG. 9 U.S. Patent 5,885,829

Lo No.te Soluble EGF (ng/ml) FIG 10A

N

Soluble EGF (ng/ml) FIG 10B U.S. Patent Mar. 23, 1999 Sheet 10 of 11 5,885,829 109

S a 108 3. vs s ds 107 s CD

106 O 20 40 60 80 100 Time (days) FIG. 11A 108

- 10 S CVO 3. e 106 CDs -- " - Collagen 105 O 20 40 60 Time (days) FIG. 11B U.S. Patent Mar 23, 1999 Sheet 11 of 11 5,885,829

3000 e E se 2000

E 2 1000

O O O 20 30 40 50 Time (days) FIG. 12 5,885,829 1 2 ENGINEERING ORAL TISSUES and compositions for use in culturing, engineering and reconstructing oral tissues. The invention generally concerns The present application claims the priority of U.S. Pro the ex vivo culture of viable oral tissue cells in combination visional Patent Application Serial No. 60/018,450 filed May with a structural matrix, or Scaffold, that results in the 28, 1996, abandoned, the entire text of which is specifically proliferation of the cells, the production of extracellular incorporated by reference herein without disclaimer. matrix, and their organization into a new tissue Structure. The regenerated oral tissue or matrix-tissue structure may BACKGROUND OF THE INVENTION then be implanted back in the body to form a new functional 1. Field of the Invention oral tissue. Methods of using Such tissueS or matrix-tissue The present invention relates generally to the fields of preparations for in Vivo drug delivery and for in Vitro dentistry and oral biology. More particularly, it concerns the toxicity and biocompatibility testing are also provided. generation of oral tissues from viable cells using ex vivo In certain embodiments, the present invention provides culture on a Structural matrix. The regenerated oral tissues methods for culturing oral tissue cells, which methods provided herein may be used in a variety of clinical appli generally comprise growing viable oral tissue cells, or cations and also in in Vitro biocompatibility testing. 15 “starter cells', on a matrix in vitro under conditions effective 2. Description of Related Art and for a period of time sufficient to allow proliferation of A major goal of dental research is the development of viable oral tissue cells. effective clinical approaches to promote the regeneration of The method may comprise growing viable oral tissue cells oral tissues following various insults or diseases. While on a three dimensional matrix in vitro under conditions Synthetic materials have been Successfully utilized as restor effective and for a period of time sufficient to allow prolif ative materials for dental tissues for a number of years eration of viable oral tissue cells to form a three dimensional (Craig, 1989), these materials do not replace the normal Structure comprising viable oral tissue cells. Alternatively, Structure and function of the lost tissue and are incapable of the method may comprise growing viable oral tissue cells on remodeling/repairing in the face of ongoing insult or Stimu a three dimensional matrix in Vitro under conditions effec lation. 25 tive and for a period of time sufficient to allow proliferation One Such example is that of root canal therapy. Approxi of viable oral tissue cells to form a three dimensional mately 15 million patients in the U.S. require a root canal biological Structure of viable oral tissue cells. each year. Here, necrotic pulp tissue resulting from trauma Further, the method may comprise growing viable oral or bacterial infection is removed and replaced with a non tissue cells on a three dimensional matrix or framework in viable Synthetic material. The Synthetic material is clearly vitro under conditions effective and for a period of time unable to provide the biological functions of pulp tissue, and sufficient to allow proliferation of viable oral tissue cells to failure leads to tooth loss. form a three dimensional Structure comprising viable oral Engineering new tissues from cultured cells represents a tissue cells on or around Said matrix or framework. Or the new approach to treat patients Suffering from the loSS or 35 method may comprise growing viable oral tissue cells on a malfunction of certain tissues (Langer and Vacanti, 1994). matrix, preferably a three dimensional matrix or Scaffold, in However, with the limited exception of oral mucosa, the vitro under conditions effective and for a period of time techniques of cell and tissue culture have not been Success sufficient to allow proliferation of viable cells that express at fully applied in the engineering of oral tissues. Current cell least one marker indicative of oral tissue cells. culture techniques, Such as those used in the regeneration of 40 The method may comprise growing viable oral tissue cells skin, and even oral mucosa, are not transferable to the on a matrix, preferably a three dimensional matrix, in Vitro regeneration of other oral tissueS as the existing techniques under conditions effective and for a period of time sufficient produce epithelia which require an appropriate connective to allow proliferation of viable cells that express one or more tissue bed in Vivo for Successful grafting. The art therefore markers indicative of oral tissue cells. Alternatively, the lackS appropriate techniques for the production of tissueS eX 45 method may comprise growing viable oral tissue cells on a Vivo that will repair and regenerate Specific oral connective matrix, preferably a three dimensional matrix, in Vitro under tissues in Vivo. conditions effective and for a period of time sufficient to Dental pulp is a loose connective tissue that provides allow proliferation of viable cells that express a plurality of dentinogenic, nutritive, Sensory and defensive functions to markers indicative of oral tissue cells. the tooth (Chiego, 1994). Dentin is produced by specialized 50 The invention also provides a method for culturing oral cells, odontoblasts, which reside in dental pulp. Tertiary tissue cells, comprising growing viable cells obtained from dentinogenesis is often initiated following injury to or loSS an oral tissue on a three dimensional matrix in vitro under of dentin by the original odontoblasts, or if lost, by uniden conditions effective and for a period of time sufficient to tified cells which differentiate into odontoblasts (Lesotet. allow proliferation of viable oral tissue cells. The method al., 1993). 55 may alternatively comprise growing viable oral tissue cells Dental pulp and other oral tissues may be capable of on a preferably three dimensional matrix in vitro under regenerating following injury, but the Specific mechanisms conditions effective and for a period of time sufficient to underlying pulp regeneration and reparative dentinogenesis allow proliferation of viable oral tissue cells in functional have not been identified. As there is little known about Such asSociation with Said matrix or Said three dimensional processes, the ability to culture or regenerate dental pulp and 60 matrix. other oral tissues has been Severely hampered. The devel The invention further provides a method for culturing oral opment of methodology by which to culture oral tissues tissue cells, comprising growing viable cells obtained from would thus represent a significant breakthrough in this field. an oral tissue in functional association with a three dimen Sional matrix in vitro under conditions effective and for a SUMMARY OF THE INVENTION 65 period of time sufficient to allow proliferation of viable oral The present invention seeks to overcome these and other tissue cells to form a three dimensional biological Structure drawbacks inherent in the prior art by providing methods comprising cells that express at least one marker indicative 5,885,829 3 4 of oral tissue cells. The invention also provides a method for in part, to the increased Surface area of the three-dimensional culturing oral tissue cells, comprising growing viable cells Support framework which results in a prolonged period of obtained from an oral tissue in functional association with a active proliferation of oral tissue cells. These proliferating three dimensional matrix in vitro under conditions effective cells elaborate proteins, Secreted extracellular matrix and for a period of time sufficient to allow proliferation of 5 components, growth factors and regulatory factors necessary viable oral tissue cells to form a three dimensional biological to Support the long term proliferation of oral tissue cells tissue structure, the cells of which tissue express at least one inoculated onto the matrix. The production of the fibrous or marker indicative of oral tissue cells. Stromal extracellular matrix tissue that is deposited on the Alternatively, the method may comprise comprising matrix is conducive for the long term growth of the oral growing viable cells obtained from an oral tissue in func tissues in Vitro. In addition, the three-dimensionality of the tional association with a three dimensional matrix in Vitro framework allows for a spatial distribution which more under conditions effective and for a period of time sufficient closely approximates conditions in Vivo for the particular to allow proliferation of viable oral tissue cells to form a oral tissues, allowing for the formation of microenviron three dimensional biological oral tissue Structure, compris ments conducive to cellular maturation and migration. The ing cells expressing at least one marker indicative of oral 15 growth of cells in the presence of this Support may be further tissue cells. Or the method may comprise growing viable enhanced by adding growth or regulatory factors, various cells obtained from an oral tissue in functional association components of extracellular matrix and other materials to with a three dimensional matrix in Vitro under conditions the Support itself or by coating the Support with these effective and for a period of time sufficient to allow prolif materials. eration of viable oral tissue cells to form a three dimensional Proliferation of cells occurs in vitro and appears to be biological oral tissue structure, the cells of which oral tissue contingent upon the geometry of the culture framework; Structure express a plurality of biological markers indicative and, when established on biodegradable framework, these of oral tissue cells. cell/matrix co-cultures are capable of regenerating oral After inoculation of the cells, the three-dimensional tissue architecture at ectopic Sites and retain their ability to framework may be incubated in an appropriate nutrient 25 Synthesize tissue-specific proteins. This cell and tissue cul medium. Many commercially available media such as RPMI ture System may have applications as a Substrate for toxicity 1640, Fisher's, Iscove’s, McCoy's, and the like may be testing and, when grown on a biocompatible or biodegrad Suitable for use. The three-dimensional oral tissue may be able polymer framework, to be implanted into Subjects. Suspended in the medium during the incubation period in Furthermore, genetically engineered cells maintain the order to maximize proliferative activity. In addition, the expression of their exogenous gene long term when grown culture may be “fed” periodically to remove the Spent media, in the culture System of the present invention. depopulate released cells, and add fresh media. The openings of the three dimensional framework or The matrices of the present invention are “structural” scaffold should be of an appropriate size to allow the cells matrices that provide a Scaffold for the cells to guide the to Stretch acroSS the openings. Maintaining actively growing process of tissue formation. Thus, the "structural matrices' 35 cells which Stretch acroSS the framework enhances the can be considered to be “three-dimensional matrices’, rather production of growth factors which are elaborated by the than Simple two dimensional Supports. It will be understood cells, and hence will Support long term cultures. In fact, any that the Scaffolding matrices of the present invention are shape or Structure of matrix that allows the cells to Stretch distinct from the inert plastic or glass ware that is used in and continue to replicate and grow for lengthy time periods routine cell culture. Not that plastic or glass is entirely 40 will work in accordance with the invention. It may be incompatible with the present invention, if modified or preferable to avoid the presence of a confluent monolayer in combined with other matrix elements to form the required the vicinity of the three-dimensional culture, as this may Scaffold-like matrix. “shut down” the growth of cells. Thus, in preferred aspects, the present invention provides 45 The invention also provides various methods for a three-dimensional, multi-layer cell and tissue culture SyS generating, or reconstructing, oral tissues. A first method tem. In particular, a culture System for the long term culture generally comprises culturing or propagating viable oral of cells and tissues in vitro in an environment that more tissue cells, or “starter cells', on a matrix in Vitro under closely approximates that found in Vivo is provided. The conditions effective and for a period of time sufficient to culture System described herein provides for proliferation 50 allow formation of an oral tissue Sample. This tissue and appropriate cell maturation to form Structures analogous approaches physiologic conditions found in Vivo to a greater to oral tissue counterparts in Vivo. The resulting oral tissues degree than other tissue culture Systems. The three Survive for prolonged periods, express oral tissue specific dimensional cell culture System is applicable to the prolif markers, perform oral tissue-specific functions, and main eration of cells and formation of tissues. tain oral tissue architecture following in Vivo implantation. 55 The “starter cells' may be enriched for a particular cell The oral tissue cultures have a variety of applications type prior to culture on the matrix. Particular cell types may ranging from transplantation or implantation in Vivo, to be selected through the use of antibodies to cell Surface Screening cytotoxic compounds and pharmaceutical com markerS Specific for a particular type of cell. Alternatively pounds in vitro, to the production of biologically active unwanted cell types may be "negatively Selected by using molecules in “bioreactors'. When grown in this three 60 antibodies specific for a marker on the Surface of the dimensional System, the proliferating oral tissue cells mature unwanted cell type. Both Selection protocols can be accom and Segregate properly to form components of oral tissues plished using monoclonal antibodies of an appropriate iso analogous to counterparts found in Vivo. type or Subclass. The invention is based, in part, on the discovery that One may obtain the composition comprising viable cells growth of oral tissue cells in three dimensions Sustains 65 from an oral tissue and immediately culture the viable cells active proliferation of oral tissue cells in culture for longer of the composition on a matrix in vitro under conditions periods of time than conventional Systems. This may be due, effective and for a period of time sufficient to allow the 5,885,829 S 6 formation of an oral tissue Sample. One may also obtain a The reconstructive methods of the present invention may composition isolated at an earlier time, or by another person, also be defined as methods which generally comprise obtain and even Stored for a moderate time period, and then culture ing a composition comprising viable cells from an oral tissue the viable cells on a matrix in vitro at a later date. Where the of an animal, culturing the viable cells of the composition on cultures are to be maintained for long periods of time or a matrix eX Vivo under conditions effective and for a period cryopreserved, non-degradable materials. Such as , of time Sufficient to allow the formation of an oral tissue dacron, polystyrene, polyacrylates, polyvinyls, teflons, Sample that resembles the corresponding native tissue , etc. may be preferred. Sample and reconstructing the oral tissue of the animal in Compositions comprising viable cells in accordance with Vivo by application of the oral tissue Sample. the present invention may be “obtained” by one party and The reconstructive methods described above may be “cultured', or otherwise manipulated as disclosed herein, by effected in Vivo by the application of a regenerated oral another party. Likewise, the regenerated tissue may be tissue Sample that has been Separated from the matrix to utilized by yet another party, either in clinical or veterinary form a regenerated oral tissue Sample Substantially free from practice or in Screening embodiments. It will therefore be matrix components prior to application of the tissue to the understood that each aspect of the present invention does not 15 animal. Alternatively, tissue reconstruction may be achieved have to be executed by the Same perSon or people, or as a by the application of a regenerated oral tissue Sample that Series of consecutive Steps without interruption. remains in combination with the matrix. The terms “culture” and “culturing”, as used herein, are Such methods are also applicable for replacing lost or preferably used to indicate culture media and techniques that damaged oral tissue, as may be caused by, e.g., an infection are known by those of skill in the art to result in successful that leads to necrotic oral tissue. An example of Such a culture of cells, preferably, mammalian cells, and most method is one which generally comprises obtaining an oral preferably, oral tissue cells. It will be understood that various tissue composition that comprises at least Some viable cells, media compositions, pH, temperature, and the like will be culturing the viable cells of the composition on a biocom Suitable. patible matrix ex vivo under conditions effective and for a It will also be understood that the use of antibiotics is 25 period of time Sufficient to allow the regeneration of an oral particularly preferred, especially as cells cultured from oral tissue Sample within the matrix and replacing the lost or tissues are typically associated with microbes, Such as yeast damaged oral tissue of the animal by implanting the matrix and bacteria, and the antibiotics will be effective to combat containing the regenerated oral tissue Sample into the lost or Such microbes. One or more antibiotics, or a cocktail damaged tissue site. thereof, will generally be used in the present invention in an An oral tissue composition that comprises Sufficient amount effective to allow the culture and growth of oral viable cells to Subsequent allow the regeneration of an oral tissue cells, without the Significant growth or proliferation of tissue may be obtained from part of a damaged tissue site microbial cells, Such as yeast and bacterial cells. The use of that is itself to be treated, or from the near Surroundings of “amounts effective' to achieve the desired results will be 35 a lost tissue. Although damaged or infected tissue may known to those of skill in the art. contain certain non-viable cells, this does not negate the In certain preferred embodiments, antibiotics will gener usefulness of the invention in culturing and regenerating oral ally be used in larger amounts than those connected with tissues from the viable cells present in Such a Sample. culture of other mammalian tissue cells. For example, up to Increased amounts of antibiotics, or cocktails thereof, should about 2 or 3 times the normal concentrations of antibiotics 40 generally be used in these contexts. and antimycotics may be used. It will also be understood that In other embodiments, an oral tissue composition that the amounts of the antibiotics or cocktails will be optimized comprises viable cells may be obtained from an oral tissue to a particular circumstance. By way of example only, Site that is compatible with, but distinct from, the tissue site where a tissue Sample from a diseased or infected tissue is to be treated. Such compatible tissues are those including the used, one of skill in the art will understand that increasing 45 Same general Structure and/or containing cells of equivalent the amounts of antibiotics employed will generally be advis regenerative capacity to the tissue site to be treated. By way able. of example only, in performing a restorative procedure on a The use of antibiotics and antimycotics in the tissue given tooth, a compatible tissue sample may be obtained culture of the present invention may be continued as long as from a distinct, healthy tooth, including an extracted tooth. desired. In this manner, the term “culturing viable cells on a 50 Viable cells can also be obtained from an oral tissue site matrix ex vivo under conditions effective and for a period of that is compatible with the site to be treated by virtue of time Sufficient to allow the formation of an oral tissue comprising cells that are capable of forming or regenerating Sample” means that the continued use, types and amounts of cells of the tissue site that is to be treated. A currently antibiotics is at the discretion of the skilled artisan carrying preferred example is that of using a Single, easily biopsied out the method as disclosed herein. 55 tissue, Such as gingiva, as the Source of cells for the in Vitro The oral tissues and viable cell compositions for use in the or eX Vivo development of oral neotissue constructs. present invention will preferably be “autologous” cells and In preferred aspects of the invention, the viable Starting tissues, which are intended for return to the animal or human cells are gingival cells obtained from a gingival tissue from which they were obtained. This approach might be Sample. In certain embodiments of the invention, the gin especially advantageous where immunological rejection of 60 gival cells are cultured to form a tissue Sample that com the transplant and/or graft verSuS host disease is likely. It is prises viable gingival Submucosal, dental pulp tissue, dentin a particular advantage of the invention that this can now be tissue, cementum tissue, periodontal tissue, oral Submucosa achieved. However, the use of other types of cells is cer tissue or tongue tissue cells. In alternate embodiments of the tainly not excluded. Therefore, “allogeneic' cells and invention, the gingival cells are cultured to form a gingival tissues, from another animal of the same Species, and even 65 Submucosal, dental pulp, dentin, cementum, periodontal, "Xenogeneic' cells and tissues, from another Species of oral Submucosa or tongue tissue Sample. In further aspects animal are contemplated for use herewith. of the invention a mixture of viable Starting cells is used. 5,885,829 7 8 In particular aspects of the invention the tissue sample cells from an oral tissue, expanding the viable cells in formed comprises viable dental pulp, dentin, gingival culture without allowing the Significant growth of microbes, Submucosa, cementum, periodontal, oral Submucosa or implanting the expanded, cultured cells onto an adherent tongue tissue cells. The tissue sample thus formed is a dental matrix in Vitro to form a matrix-cell preparation and main pulp, dentin, gingival Submucosa, cementum, periodontal, taining the preparation under conditions effective and for a oral Submucosa or tongue tissue Sample. period of time sufficient to allow cell proliferation and The tissue sample may be formed by culturing viable organization into an oral tissue Sample that resembles, e.g., Starting cells obtained from an oral tissue Sample enriched in Structurally and functionally, the corresponding native tissue dental pulp-derived fibroblasts. In certain aspects of the Sample. invention the viable Starting cells enriched in dental pulp In terms of regenerating oral tissue for use in tissue derived fibroblasts are obtained from an extracted tooth. In reconstruction, the present invention also provides methods particular embodiments of the present invention, the viable which generally comprise obtaining a composition that Starting cells enriched in dental pulp-derived fibroblasts are comprises at least Some viable cells from an oral tissue of an obtained from a tissue Sample extracted from a tooth that animal, culturing the viable cells, preferably in the presence remains in an animal. Additionally, the tissue sample may be 15 of an amount of an antimicrobial or antibiotic effective to formed by culturing viable Starting cells obtained from an inhibit the growth of microbes, Seeding the cultured cells on oral tissue Sample enriched in gingival Submucosal fibro a matrix to form an eX Vivo matrix-cell preparation, cultur blasts. ing the matrix-cell preparation under conditions effective In other aspects of the invention the tissue sample formed and for a period of time sufficient to allow proliferation and may comprise viable dentin, gingival Submucosa, organization of the cells to form an oral tissue Sample cementum, periodontal, oral Submucosa or tongue tissue asSociated with the matrix and implanting the cultured oral cells. Thus, the tissue Samples formed may be dentin, tissue Sample into an oral tissue site of the animal to effect gingival Submucosa, cementum, periodontal, oral Submu reconstruction of the oral tissue. cosa or tongue tissue Sample. In these aspects, the tissue It will be understood that the oral tissue Sample may again Samples may be formed by culturing viable starting cells 25 be separated from the matrix prior to application to the obtained from an oral tissue Sample enriched in dental pulp, animal. Equally, the oral tissue Sample may be applied to the gingival Submucosal fibroblast, periodontal ligament or oral animal in combination with the matrix, wherein the matrix Submucosal fibroblast cells. would preferably be a biocompatible matrix. Implantation of In further embodiments of the invention the tissue sample a cultured matrix-cell preparation into a Specific oral tissue formed may comprise viable oral tissue cells of at least two Site of an animal to effect reconstruction of oral tissue may different cell types. In other embodiments of the invention involve a biodegradable matrix or a non-biodegradable the tissue sample formed may comprise a plurality of matrix, depending on the intended function of the prepara distinct viable oral tissue cells. tion. Gingival Samples are contemplated for use in the present The present invention may also be used in connection invention for the regeneration of dental pulp, dentin, peri 35 with other Standard techniques of dentistry and oral medi odontal tissue, and bone, as well as oral Submucosa and cine. The local or Systemic administration of antimicrobials gingival Submucosa (Subgingival connective tissue). Using a in connection with the tissue administration described herein gingival tissue Sample means that a tooth does not need to is also contemplated. be extracted in order to obtain pulp or periodontal ligament 40 Although by no means required, in certain embodiments fibroblasts as a Source of cells. Gingival biopsies are obtain the oral cell and tissue culture methods of the present able by routine dental procedures with little or no attendant invention may utilize certain factors that regulate the growth donor site morbidity. and/or the function of oral tissue cells. Such exogenous The use of gingival cells is currently preferred for the factors may be used in the context of cell or tissue culture eX induction of mineralized oral connective tissues, Such as 45 vivo and/or in the modulation of cell or tissue function in dentin, cementum and bone. However, the generation of Vivo, following delivery to the animal as part of the reim muscle cells as part of the tongue regeneration is not plantation process. excluded as the gingival fibroblasts may be engineered to Growth factors and regulatory factors need not be added differentiate into muscle cells. to the media since these types of factors are elaborated by In certain preferred embodiments, the invention concerns 50 the three-dimensional cells. However, the addition of Such the generation of a matrix-cell preparation and the “culture' factors, or the inoculation of other specialized cells may be or maintenance of Such a preparation for a period of time used to enhance, alter or modulate proliferation and cell before implantation or further use, during which time further maturation in the cultures. The growth and activity of cells cell proliferation and tissue regeneration occurs. in culture can be affected by a variety of growth factorS Such Accordingly, the present invention also provides methods 55 as insulin, growth hormones, Somatomedins, colony Stimu for generating an oral tissue which generally comprise lating factors, erythropoietin, cytokines and other growth obtaining a composition that comprises Sufficient numbers factors. Other factors which regulate proliferation and/or of viable cells from an oral tissue, culturing the cells of the differentiation include prostaglandins and interleukins. composition, preferably using antibiotics or cocktails Accordingly, the present invention further provides cell thereof, implanting the cells on a matrix in Vitro to form a 60 culture methods where viable oral tissue cells are cultured in matrix-cell preparation and maintaining the matrix-cell the presence of one or more exogenous factors that Stimulate preparation under conditions effective and for a period of the growth or proliferation of the cells. Further, the cells may time Sufficient to allow the formation of an oral tissue be cultured on a matrix in the presence of at least one Sample. exogenous factor that Stimulates the growth or proliferation These methods may also be further defined as methods for 65 of the cells on the matrix. The cells, tissues and matrix generating oral tissue which generally comprise isolating an tissues may also be cultured and/or maintained in the oral tissue composition that comprises at least Some viable presence of one or more exogenous factors that inhibit 5,885,829 9 10 adverse processes, Such as those factors that inhibit cell uct. Secondly, gene therapy techniques are useful only if the death and apoptosis. Any Such factor or factors may be number of transfected cells can be Substantially enhanced to Semi-purified, purified, natural or recombinant. be of clinical value, relevance, and utility; the three dimensional cultures of the invention allow for expansion of In another embodiment of the invention the viable oral the number of transfected cells and amplification (via cell tissue cells are provided with an exogenous gene that division) of transfected cells. Preferably, the expression expresses an exogenous factor in the cells. The exogenous control elements used should allow for the regulated expres gene may be provided on a recombinant vector comprising Sion of the gene So that the product is Synthesized only when the exogenous gene encoding the exogenous factor, wherein needed in Vivo. The promoter chosen would depend, in part the recombinant vector expresses the exogenous gene in the upon the type of tissue and cells cultured. Cells and tissues cells. The exogenous gene may be provided to the cells prior which are capable of Secreting proteins are preferable. to Seeding the cells on the matrix, towards the beginning of In the context of tissue application in Vivo, regenerated the in vitro matrix culture process or towards the end of the oral tissue may be applied to an oral tissue site of an animal in vitro matrix culture process. in the presence of one or more exogenous factors that In certain embodiments of the invention the exogenous 15 Stimulate the growth or proliferation of the tissue in the gene expresses an exogenous factor that Stimulates the animal, and/or cells that naturally produce Such factors growth or proliferation of the cells. In other aspects, the and/or cells that have been engineered to produce Such exogenous gene expresses an exogenous therapeutic factor factors. Further, a biocompatible matrix containing a regen that is released by the cells. A wide variety of exogenous erated oral tissue Sample may be implanted into a lost or therapeutic factors are contemplated for use in the present damaged tissue site of an animal in the presence of one or invention, as exemplified by antibiotics, growth factors, more exogenous factors or cells that Stimulate the growth or cytokines, blood clotting factors and insulin. The use of a proliferation of the tissue in the animal, or promotes vascu particular therapeutic agent will be dependent on the con larization of the tissue or effects other beneficial results. dition in the animal that is to be treated, and are generally well known to those of skill in the art. It can thus be seen that the present invention provides 25 methods for delivering factors, Such as growth factors, to In further embodiments, the exogenous factor or factors oral tissues within an animal, which methods generally used to further stimulate the growth or proliferation of the comprise culturing a composition that comprises at least cells in culture (or to inhibit cell death), either before or Some viable oral tissue cells on a matrix in vitro under during the matrix culture phase, may be an exogenous conditions effective and for a period of time sufficient to growth or proliferation factor produced by a cell or popu allow formation of an oral tissue Sample and delivering the lation of cells that is co-cultured with the cells and/or tissues Sample to the oral tissue site in the presence of an exogenous to be regenerated. The present invention thus contemplates factor. Such methods may also be generally defined as the use of natural cells that elaborate growth factors, including the steps of obtaining at least Some viable cells hormones, cytokines, and other autocrine, paracrine, hor from an oral tissue, culturing the cells on a biocompatible monal and neurotransmitter-like Stimulants in the culture 35 matrix in vitro under conditions effective and for a period of and matrix-culture phases of oral tissue regeneration. time Sufficient to allow formation of a cultured matrix-cell In Still further embodiments, the growth factor, hormone, preparation, admixing the matrix-cell preparation with at cytokine, hormone, neurotransmitter, cell death inhibitor, or least one Selected factor and applying the matrix-cell like molecule may be released into the cell, tissue or Selected factor admixture to an oral tissue site of an animal. matrix-tissue culture by a recombinant cell engineered to 40 The invention further provides methods for delivering produce the growth factor, hormone, cytokine, hormone, Stimulatory and growth factors to oral tissues within an neurotransmitter or desired molecule. The exogenous factor animal, which methods generally comprise culturing a com may be produced by a recombinant cell by providing the cell position that comprises at least Some viable oral tissue cells with the exogenous factor on a recombinant vector that on a matrix in Vitro under conditions effective and for a expresses the factor in the recombinant cell. Recombinant 45 period of time Sufficient to allow formation of an oral tissue engineering for protein production and Secretion is now Sample and delivering the Sample to the oral tissue site in the routine in the art. presence of a natural cell, a recombinant cell or a population Thus, the three-dimensional culture System of the inven of cells that produce Such factors. Biocompatible matrices tion may afford a vehicle for introducing genes and gene may also be used to prepare a matrix-cell preparation which products in Vivo for use in gene therapies. For example, 50 is admixed with a native or recombinant cell that produces using recombinant DNA techniques, a gene for which a at least one Selected factor and applied to an oral tissue site patient is deficient could be placed under the control of a of an animal. viral or tissue-specific promoter. The recombinant DNA A wide variety of Structural matrices may be used in the construct containing the gene could be used to transform or context of the present invention. In certain embodiments, the transfect a host cell which is cloned and then clonally 55 matrices will preferably be biocompatible matrices. expanded in the three-dimensional culture System. The Naturally, any embodiment that concerns the application of three-dimensional culture which expresses the active gene a matrix, or Substantial portions thereof, to an oral tissue site product, could be implanted into an individual who is will require the matrix to be Such a biocompatible matrix. deficient for that product. The engineered oral tissues would Where in Vivo application of a matrix-tissue Sample is thus act as an “implantable pump', for the in Vivo produc 60 contemplated, the biocompatible matrix may be a biode tion and Secretion of a desired gene product. Additionally, gradable matrix, a slowly biodegradable matrix or a non should the need arise to halt production of the exogenous biodegradable matrix, depending on the particular tissue and gene, the tissue could simply be removed from the animal. clinical application. The use of non-biodegradable matrices The use of the three-dimensional culture in gene therapy is routine in dentistry and will be Suitable for certain cases. has a number of advantages. Firstly, Since the culture com 65 The use of biodegradable compounds in clinical Situations is prises eukaryotic cells, the gene product will be properly equally established, and is contemplated for use in other expressed and processed in culture to form an active prod embodiments of the invention. 5,885,829 11 12 In contrast, other embodiments of the present invention als are used as the three-dimensional Support framework, it do not require the use of a biocompatible matrix. For may be advisable to pre-treat the matrix prior to inoculation example, where the method involves the complete or Sub of cells in order to enhance the attachment of cells to the Stantial degeneration of the matrix prior to application of the framework. For example, prior to inoculation with cells, cultured tissue sample to an animal, the biocompatibility of nylon screens could be treated with 0.1M acetic acid, and the initial Starting matrix is not particularly important. incubated in polylysine, FBS, and/or collagen to coat the Similarly, where the method involves the removal or “har nylon. Polystyrene could be similarly treated using Sulfuric Vest' of regenerated tissue from the matrix, the biocompat acid. Where the three-dimensional culture is itself to be ibility of the matrix is largely irrelevant. In the former implanted in Vivo, it may be preferable to use biodegradable Situation, the matrix may be both generally unstable, labile materials. Such as PGA, Suture material, collagen, or chemically degradable, and also non-biocompatible. In , or hyaluronic acid. For example, these the latter case, a non-biocompatible matrix may be either materials may be woven into a three-dimensional framework degradable or non-degradable, So long as the tissue can be Such as a collagen Sponge. isolated substantially free from the non-biocompatible ele In other embodiments, matrices for use in the invention ments of the matrix prior to administration to the body. 15 may be naturally-derived matrices or “biomatrices”. Pre Preferred matrices for use in the present invention will ferred examples of biomatrices are those extracted from or generally be those that define a Space for the Subsequent resembling the extracellular matrix (ECM). These matrices tissue development. Such matrices include hydrogels, or may be termed ECM polymer matrices. One currently porous matrices Such as fiber-based or Sponge-like matrices. preferred example of a naturally-derived, or “ECM matrix”, In certain embodiments, Synthetic matrices, as exempli is a collagen matrix, Such as type I collagen. fied by Synthetic polymer matrices, may be used. The Further suitable examples of naturally-derived matrices matrices may be homopolymers or heteropolymerS. Certain include laminin-rich gels, alginate, agarose and other examples of Such synthetic matrices are polylactic acid polysaccharides, gelatin, fibrin glues, and hyaluronic acid (PLA) polymer matrices, polyglycolic acid (PGA) polymer derivatives. Any Such matrix, and blends of these materials matrices and polylactic acid-polyglycolic acid (PLGA) 25 with other polymers or other materials, is contemplated for copolymer matrices. Both the Stereoisomeric forms are use in the present invention. contemplated to be useful. Chemically, these may also be In certain embodiments, “Second generation matrices” or termed poly-(L-lactic acid), PLA or PLLA, and poly-(D.L- Second generation ECM matrices may be used. These are lactic acid), PDLLA. PLGA may also be written poly-(D, exemplified by Synthetic polymer matrices that have L-lactic-co-glycolic acid). appended, i.e., are operatively attached to, one or more Such PLA, PGA and PLGA matrices may take various biologically active molecules. Such biologically active mol forms, including fiber matrices, tubular matrices and Sponge ecules include Saccharides, polysaccharides, amino acids, matrices. The Sponge matrices may include PLA and poly peptides, polypeptides, proteoglycans, glycoproteins, and vinyl alcohol (PVA). the like. Thus, these matrices have functions of both natural Further examples of appropriate Synthetic matrices are 35 and Synthetic matrices. They may include the products of polyanhydrides, , polyorthoesters, and poly(amino Synthetic chemistry and recombinant protein production. acids), polypeptides, oxide, The present invention is generally applicable to the cul polyphosphaZenes, various block copolymers, Such as those ture and regeneration of a variety of oral tissues and struc consisting of ethylene oxide and propylene oxide (e.g., tures. By way of example only, one may mention dental pulp Pluronic surfactant; BASF Corp.), and blends of polymers 40 tissue, dentin, periodontium, bone, cementum, gingival from this group and with other polymers. Further polymers Submucosa, oral Submucosa, Salivary gland tongue and taste are detailed herein in Example IV. Ceramics, Such as cal bud tissues. cium phosphate matrices, may also be employed in the A currently preferred application of the present invention present invention. 45 is in the culture and regeneration of dental pulp tissue. In The three-dimensional Support framework may be of any Such embodiments, dental pulp tissue is generally regener material and/or shape that allows cells to attach to it (or can ated by culturing gingival tissueS or dental pulp-derived be modified to allow cells to attach to it), and allows cells to cells, such as fibroblasts. However, the culture of other cells grow in more than one layer. A number of different materials within an extracted dental pulp tissue, Such as Odontoblasts, may be used to form the framework, including but not 50 may also contribute to the regeneration of dental pulp tissue limited to: nylon (polyamides), dacron (polyesters), according to the present invention. polystyrene, polypropylene, polyacrylates, polyvinyl com In certain embodiments, the tissue for use in the regen pounds (e.g., polyvinylchloride), polycarbonate (PVC), eration process will be obtained from the gingiva. Gingival polytetrafluorethylene (PTFE; teflon), thermanox (TPX), Samples are readily obtainable by routine biopsy. , cotton, polyglycolic acid (PGA), cat gut 55 In other embodiments, an original Sample of dental pulp Sutures, , gelatin, dextran, etc. Any of these mate tissue for use in the regeneration process will be obtained rials may be woven into a , for example, to form the from a healthy tooth, Such as an extracted molar or wisdom three-dimensional framework. tooth. The matrix may be composed of individual matrix In further embodiments, the Sample of dental pulp tissue components, and the individual matrix components may be 60 containing the Starter cells for regeneration will be obtained, coated with a coating agent. In further aspects of the or Salvaged, from the tooth to be operated on. In the latter invention the matrix is a Synthetic fiber matrix, the indi process, the biological material isolated from a given tooth vidual fiber components of which matrix are coated with a will be used to regenerate healthy tissue from the viable cells collagen coating agent, a polylysine coating agent or an FBS that exist in the extracted Sample, notwithstanding the fact coating agent. 65 that certain non-viable cells will likely be present in Such a Certain materials, Such as nylon, polystyrene, etc., may be Sample. The existence of certain non-viable cells, and/or poor substrates for cellular attachment. When these materi cells not of oral tissue origin, in a Sample extracted from a 5,885,829 13 14 diseased or damaged tooth does not generally negate the In a particular embodiment of the invention, the viable operativity of the tissue regeneration methods of the present Starting cells are obtained from a gingival tissue sample. The invention. The determination of whether a given Sample is viable oral cells of the present invention may form a three appropriate for use in tissue regeneration will be Straight dimensional biological Structure. In certain aspects, the forward to those of skill in the art in light of the present viable oral cells form a three dimensional oral tissue struc disclosure. Using tissue samples obtained from a single ture. In a further embodiment of the invention, the three tooth has the advantage that a distinct healthy tooth does not dimensional biological or oral tissue structure resembles the have to be invaded in order to obtain the viable cells. corresponding native oral tissue. It will be understood by those of ordinary skill in the art Additionally, the present invention provides a method for that the regeneration of a particular oral tissue from a reconstructing an oral tissue or for replacing lost or damaged Starting tissue Sample will require a Sample to be obtained oral tissue in an animal, comprising obtaining a composition that comprises viable cells capable of regenerating the comprising viable starting cells from an oral tissue Sample of particular oral tissue desired. an animal, culturing the viable Starting cells of Said com The use of gingival Samples is contemplated to be advan position on a matrix eX Vivo under conditions effective and tageous as this can give rise to regenerated tissueS of dental 15 for a period of time sufficient to allow the formation of a pulp, dentin, periodontium, cementum, bone, oral tissue sample that comprises oral tissue cells that resemble Submucosa, gingival Submucosa, and even has the potential the corresponding native oral tissue cells and applying Said to regenerate Striated muscle cells. tissue sample to an oral tissue site in Vivo to reconstruct the oral tissue or to replace the lost or damaged oral tissue of the In regard to other exemplary tissues: animal. to culture and regenerate dentin tissue, one would obtain The viable Starting cells may be obtained from a gingival a Sample containing dental pulp cells or gingival Submucosal tissue Sample. Alternatively, the viable Starting cells may be fibroblasts; obtained from an oral tissue Sample that is obtained from an to regenerate periodontal tissue, one would generally oral tissue site compatible with, but distinct from, the oral obtain a tissue Sample containing periodontal ligament cells 25 tissue to be reconstructed or replaced. The viable starting or gingival Submucosal fibroblasts, cells may also be obtained from an oral tissue Sample that is to culture and regenerate gingival Submucosal tissue obtained from the damaged oral tissue site to be recon (Subgingival connective tissue), one would generally use a Structed or replaced. Sample containing gingival Submucosal fibroblasts, In a particular aspect of the invention, the viable Starting to regenerate an oral Submucosal tissue, a tissue sample cells are cultured on the matrix in Vitro in the presence of an comprising oral Submucosal fibroblasts would be used; and amount of an antimicrobial agent effective to inhibit the to generate a tongue tissue Sample, one would generally growth of microbial cells in the culture. In another embodi use a tissue Sample containing gingival Submucosal fibro ment of the invention, the viable starting cells are pre blasts. 35 cultured in vitro for a period of time effective to prepare an Dental pulp, oral and gingival Submucosal tissues are expanded population of viable cells for culture on Said advantageous as they are constitutively capable of tissue matrix in vitro. regeneration. However, the use of viable cells isolated from The tissue samples of the invention may be separated the oral or gingival Submucosa in the regeneration of other from the matrix prior to application to the animal, or may be tissues, Such as dental pulp, dentin, cementum, periodontal 40 applied to the animal in combination with the matrix. ligament and bone, will be understood to involve, in certain In a particular aspect of the invention, the tissue Sample embodiments, the induction of particular developmental has a Structure that resembles the corresponding native oral pathways by, e.g., the addition of Specific protein factors or tissue structure. In an additional aspect of the invention the exposure to particular conditions. Genetic engineering to tissue Sample is applied to the oral tissue site to be recon induce, promote or assist in the development of a particular 45 Structed or replaced in the presence of an exogenous factor tissue is also contemplated. that Stimulates the growth or proliferation of the tissue in the The invention further provides a method for culturing oral animal. In a further aspect of the invention, the exogenous tissue cells, comprising growing viable starting cells factor is produced by a cell that is also applied to the oral obtained from an oral tissue sample in functional association tissue site of Said animal. In Still further aspects of the with a three dimensional matrix in Vitro under conditions 50 present invention, the exogenous factor-producing cell is a effective and for a period of time sufficient to allow prolif recombinant cell engineered to produce Said factor. eration and organization of viable oral tissue cells to form a Alternatively, the exogenous factor-producing cell may be a three dimensional biological Structure comprising cells that cultured oral tissue cell of the applied tissue Sample, the cell express at least one marker indicative of oral tissue cells. In being provided with an exogenous gene that expresses Said particular embodiments of the invention the viable oral cells 55 exogenous factor. In certain aspects, the cell is provided with are analyzed to confirm the presence of at least one biologi a recombinant vector that comprises the exogenous gene. cal marker indicative of oral tissue cells. In a particular embodiment of the invention, the oral The invention also provides a method for generating an tissue cells of the tissue Sample are provided with an oral tissue Sample, comprising growing viable starting cells exogenous gene that expresses an exogenous factor in the obtained from an oral tissue sample in functional association 60 cells prior to application of Said tissue sample to Said animal. with a three dimensional matrix in Vitro under conditions In various aspects of the invention, the exogenous gene is effective and for a period of time sufficient to allow prolif provided to the cells prior to the culture of the cells on the eration and organization of viable oral tissue cells to form a matrix, towards the beginning of the in Vitro matrix culture three dimensional oral tissue Structure comprising a popu process, or towards the end of the in Vitro matrix culture lation of cells that comprise at least one Sub-population of 65 proceSS. cells that express one or more biological markers indicative In one aspect of the invention, the exogenous gene of oral tissue cells. expresses an exogenous factor that Stimulates the growth or 5,885,829 15 16 proliferation of the oral tissue cells of the tissue Sample tooth in Step (a), which tissue comprises at least Some viable when applied to the animal. In another aspect of the dental pulp cells. In a first alternative, the tissue Sample invention, the exogenous gene expresses an angiogenic comprising at least Some viable cells capable of regenerating factor that stimulates the growth or proliferation of blood into dental pulp cells may be obtained from a distinct, vessels in or around the oral tissue cells of the tissue Sample healthy tooth, Such as a molar or wisdom tooth of the animal, following application to the animal. In a further aspect of the which may tooth may be extracted for the purposes of invention, the exogenous gene expresses a factor that inhib obtaining the necessary Sample. In a further alternative, the tissue sample obtained in Step (b) may be obtained from a its apoptosis in the oral tissue cells of the tissue Sample gingival Sample comprising gingival Submucosal fibro following application to the animal. blasts. In yet further aspects, the exogenous gene expresses an In certain exemplary embodiments, the methods for filling exogenous therapeutic factor, Such as an antibiotic, a growth a root canal may be defined as those which comprise the factor, a hormone, a cytokine, a blood clotting factor or Steps of insulin, that is released by the cells of the tissue following a) removing dental pulp tissue, Such as infected dental application to the animal. pulp tissue, from the root of a diseased or damaged In certain exemplary embodiments, the present invention 15 tooth of an animal to create a cleansed root chamber; provides methods and compositions for filling root canals. b) placing a temporary implant in said root chamber; One Such method generally comprises obtaining dental pulp c) obtaining viable tissue comprising at least Some cells tissue comprising at least Some viable dental pulp cells from capable of regenerating into dental pulp tissue, i.e., an animal, either from the tooth to be treated, from a distinct, “viable dental pulp-regenerative tissue', Such as a healthy tooth, or from gingival Submucosal fibroblasts, dental pulp tissue Sample from a tooth of the animal, culturing the cells of the tissue on a matrix eX Vivo, generally e.g., a healthy molar or wisdom tooth of the animal, or using antimicrobials, and under conditions effective and for a gingival Sample comprising gingival Submucosal a period of time Sufficient to allow the regeneration of dental fibroblasts; pulp tissue in connection with the matrix; and implanting the d) culturing viable fibroblast cells from the tissue of step regenerated dental pulp tissue into the tissue Space of the 25 (c) in vitro, generally using an effective amount of at root canal. least one antimicrobial; In a particular embodiment of the invention, the Starting e) implanting the cultured cells from Step (d) on a bio oral tissue Sample is enriched in dental pulp-derived fibro compatible matrix to form a matrix-cell preparation; blasts. In certain aspects of the invention, the Starting oral f) maintaining the matrix-cell preparation of Step (e) tissue Sample is obtained from the dental pulp tissue of an under conditions effective and for a period of time extracted tooth. In alternative aspects, the Starting oral tissue Sufficient to allow the formation of matrix-tissue prepa Sample is obtained from a dental pulp tissue Sample obtained ration containing regenerated dental pulp tissue; from a tooth that remains in an animal. In other aspects, the g) admixing the matrix-tissue preparation of Step (f) with dental pulp tissue Sample is obtained from a healthy molar 35 an exogenous factor that Stimulates the growth of or wisdom tooth of Said animal distinct from the target tooth. proliferation of oral tissue to create a matrix-tissue In yet other embodiments, the dental pulp tissue Sample is obtained from the target tooth to be treated. In particular factor admixture; aspects, the Starting oral tissue Sample is enriched in gingival h) removing the temporary implant of step (b) from the Submucosal fibroblasts. root chamber of step (a); and 40 i) implanting the matrix-tissue factor admixture of step (g) The regenerated dental pulp tissue may be separated into the root chamber re-created in Step (h). distinct from the matrix prior to application into the tissue The invention further provides a method for delivering a Space of the root canal or it may be applied to the tissue Selected factor to an animal, comprising culturing viable oral Space of the root canal in combination with the matrix, tissue cells on a matrix in vitro under conditions effective wherein the matrix is a Substantially biocompatible matrix. 45 and for a period of time sufficient to allow formation of a Such methods may also be defined as those which gen tissue Sample that comprises viable oral tissue cells and erally comprise the Steps of delivering Said tissue Sample to an oral tissue site of an a) removing dental pulp tissue, Such as infected dental animal in the presence of a Selected factor. The method may pulp tissue, from the root of a diseased or damaged be characterized as comprising the Steps of culturing viable tooth of an animal to create a root chamber; 50 oral tissue cells on a matrix in Vitro under conditions b) obtaining a tissue sample comprising at least Some effective and for a period of time sufficient to allow forma viable cells capable of regenerating into dental pulp tion of a tissue Sample that comprises viable oral tissue cells, cells, e.g., a dental pulp Sample from a tooth of the admixing the oral tissue cells with the Selected factor, and animal, or a Sample containing gingival Submucosal applying the admixture of oral tissue cells and the Selected fibroblasts, and culturing the tissue Sample on a bio 55 factor to the oral tissue site of the animal. compatible matrix eX Vivo, optionally in the presence In another aspect of the invention, the Selected factor is of an exogenous factor that Stimulates the growth or produced by a cell and the cell is delivered to the oral tissue proliferation of the tissue in the matrix, and under Site in the presence of the tissue Sample. The method may be conditions effective and for a period of time sufficient characterized as comprising the Steps of culturing viable oral to allow the formation of a matrix-tissue preparation 60 tissue cells on a matrix in vitro under conditions effective containing regenerated dental pulp tissue, and and for a period of time sufficient to allow formation of a c) implanting isolated regenerated dental pulp tissue, or a tissue sample that comprises viable oral tissue cells, admix combined matrix-regenerated dental pulp tissue ing the oral tissue cells with a Second population of cells that preparation, into the root chamber created in Step (a). produces the Selected factor, and applying the admixture of The tissue Sample comprising at least Some viable cells 65 oral tissue cells and the Second population of cells that capable of regenerating into dental pulp cells obtained in produce the Selected factor to the oral tissue Site of the Step (b), above, may be dental pulp tissue obtained from the animal. 5,885,829 17 18 In a further embodiment of the invention, the selected ized as being regenerated and having a structure, generally factor is produced by a recombinant cell engineered to a three-dimensional Structure, that corresponds to the natural produce the factor and the recombinant cell is delivered to tissue. The oral tissue Samples may be further characterized the oral tissue site in the presence of Said tissue sample. The as those prepared by a process comprising culturing a method may be characterized as comprising the Steps of composition comprising at least Some viable oral tissue cells culturing viable oral tissue cells on a matrix in vitro under on a matrix in Vitro under conditions effective and for a conditions effective and for a period of time sufficient to period of time Sufficient to allow formation of an oral tissue allow formation of a tissue Sample that comprises viable oral Sample. tissue cells, obtaining a recombinant cell engineered to The regenerated dental pulp, dentin, periodontium, gin produce the factor, admixing the oral tissue cells and the gival Submucosa, oral Submucosa or tongue tissueS of the recombinant cell engineered to produce the factor, and invention may be further defined as those regenerated oral delivering the admixture of oral tissue cells and the recom tissue samples prepared by a process that comprises cultur binant cell engineered to produce the factor to the oral tissue ing a composition comprising viable oral tissue cells on a Site of the animal. matrix in vitro to form a cultured matrix-cell preparation and In yet other embodiments, the Selected factor is produced 15 maintaining the matrix-cell preparation in the presence of at by an oral tissue cell of the delivered tissue Sample, the oral least one exogenous factor, which may be a purified factor, tissue cell being provided with an exogenous gene that or a factor produced by a natural cell, or a recombinant cell, expresses Said Selected factor. The method may be charac wherein the factor stimulates the growth or proliferation of terized as comprising the Steps of culturing viable oral tissue the cells and under conditions effective and for a period of cells on a matrix in vitro under conditions effective and for time Sufficient to allow the formation of a regenerated oral a period of time Sufficient to allow formation of a tissue tissue Sample. Sample that comprises viable oral tissue cells, providing an In still further embodiments, this invention provides exogenous gene encoding the Selected factor to the oral matrices that are associated with or that contain, i.e., tissue cells, and administering the oral tissue cells provided “house', a cultured or regenerated oral tissue Sample. In with the exogenous gene encoding the Selected factor to the 25 certain embodiments, the matrix will be a biocompatible oral tissue site of the animal. In a particular aspect of the matrix, allowing administration of the tissue-containing invention, the exogenous gene is provided to the oral tissue matrix to an oral tissue site of an animal. It may also be a cells prior to culturing on the matrix. Slowly biodegradable matrix. In a particular aspect of the invention the Selected factor The methods and matrix-tissue preparations of the present Stimulates the growth or proliferation of the oral tissue cells invention are advantageous in that they allow the generation of the tissue Sample delivered to Said animal. In another of a three dimensional oral tissue Sample, rather than a two aspect of the invention, the Selected factor Stimulates the dimensional layer of cultured cells. Accordingly, the inven growth or proliferation of blood vessels in or around the oral tion provides three dimensional matrices, including biocom tissue cells of the tissue sample delivered to the animal. In patible matrices, that are associated with or that house a further aspect of the invention, the Selected factor provides 35 cultured or regenerated oral tissue Samples prepared by a an oral therapeutic benefit upon delivery to an oral tissue site process which generally comprises obtaining a composition of an animal. In Still other aspects of the invention, the including at least Some viable oral tissue cells and culturing Selected factor provides a Systemic therapeutic benefit upon the cells on a biocompatible matrix in Vitro under conditions delivery to an oral tissue site of an animal and Subsequent effective and for a period of time sufficient to allow prolif uptake into the Systemic circulation of the animal. In a 40 eration and organization of the cells to form an oral tissue particular aspect of the invention, the Selected factor is Sample associated with or housed within the matrix. Subsequently removed from the animal by removing the In certain embodiments, the matrix-tissue preparations of delivered tissue Sample from the oral tissue site of the the invention are those that are associated with or house a animal. regenerated oral tissue Sample prepared by a process com The invention also provides various compositions which 45 prising obtaining a viable oral tissue cell composition and may be used in the context of tissue regeneration in Vivo and culturing the cells on a matrix in vitro to form a cultured also in a number of in vitro Screening embodiments. A first matrix-cell preparation and maintaining the matrix-cell composition in accordance with the present invention is a preparation in the presence of an exogenous factor that population of cultured oral tissue cells, Substantially free Stimulates the growth or proliferation of the cells and under from microbial cells, which oral tissue cells are prepared by 50 conditions effective and for a period of time sufficient to a process generally comprising growing a composition com allow proliferation and organization of the cells to form an prising at least Some viable oral tissue cells on a matrix in oral tissue Sample within the matrix. vitro under conditions effective and for a period of time In currently preferred embodiments, the present invention sufficient to allow proliferation of viable oral tissue cells. provides polylactic acid (PLA) polymer, polyglycolic acid Further populations of cells in accordance with this inven 55 (PGA) polymer or polylactic-polyglycolic acid (PLGA) tion are those Substantially microbe-free populations of cells co-polymer matrices in combination with a regenerated prepared by processes comprising growing compositions dental pulp tissue Sample. Further, currently preferred com comprising at least Some viable oral tissue cells on a matrix positions are those comprising PLA, PGA or PLGA fiber or in vitro in the presence of at least one exogenous factor that Sponge matrices associated with, or housing, a regenerated Stimulates the growth or proliferation of the cells and under 60 dental pulp tissue sample, prepared by a proceSS comprising conditions effective and for a period of time sufficient to culturing viable fibroblasts from a dental pulp or gingival allow proliferation of viable oral tissue cells. submucosal sample on a PLA, PGA or PLGA matrix in vitro In further embodiments, the invention provides various under conditions effective and for a period of time sufficient regenerated, Substantially microbe-free, oral tissue Samples to allow proliferation and organization of the fibroblasts to including dental pulp, dentin, periodontium, gingival 65 form a dental pulp tissue Sample associated with the matrix. Submucosa, oral Submucosa and tongue tissue Samples. The The invention further provides a method for testing the oral tissue samples of the invention are generally character Suitability of a candidate Substance for use in the oral cavity, 5,885,829 19 20 comprising applying Said candidate Substance to a Vitro to Screen cytotoxic and/or pharmaceutical compounds regenerated, three-dimensional oral tissue Sample and ana in order to identify those that are most efficacious, i.e., those lyzing the effect of said substance on the viability of said that kill the malignant or diseased cells, yet spare the normal tissue sample. The three-dimensional liver cultures may be cells. These agents could then be used to therapeutically used in vitro to Screen a wide variety of compounds, Such as treat the patient. cytotoxic compounds, growth/regulatory factors, pharma In Still further embodiments, the present invention pro ceutical agents, etc. To this end, the cultures are maintained vides oral or dental kits comprising regenerated tissues, or in vitro and exposed to the compound to be tested. The matrix-tissue preparations, in accordance with the present activity of a cytotoxic compound can be measured by its invention. The kits may generally be defined as those ability to damage or kill cells in culture. This may readily be comprising, in Suitable container means, a regenerated oral assessed by Vital Staining techniques. The effect of growth/ tissue sample, or a three dimensional matrix associated with regulatory factors may be assessed by analyzing the cellular or housing a regenerated oral tissue Sample. The kits of the content of the matrix, e.g., by total cell counts, and differ invention include both therapeutic kits and test or assay kits. ential cell counts. This may be accomplished using Standard The therapeutic kits provide tissues and biocompatible cytological and/or histological techniques including the use 15 matrix-tissue preparations for administration to an animal or of immunocytochemical techniques employing antibodies human Subject. The test kits are generally intended for use that define type-specific cellular antigens. The effect of in conducting in vitro assays to determine whether a candi various drugs on normal cells cultured in the three date Substance is Suitable for use in the oral cavity. dimensional System may be assessed. In yet still further embodiments, the invention thus pro The three-dimensional oral tissue culture System of the vides methods for testing the Suitability of a candidate invention can be used in a variety of applications. These Substance for use in the oral cavity, comprising applying a include but are not limited to transplantation or implantation candidate Substance to an isolated regenerated oral tissue or of the cultured cells in Vivo, Screening cytotoxic to a three dimensional matrix associated with a regenerated compounds, carcinogens, mutagens growth/regulatory oral tissue sample and analyzing the effect of the Substance factors, pharmaceutical compounds, etc., in vitro, elucidat 25 on the viability of the tissue sample. It will be readily ing the mechanism of certain diseases, Studying the mecha understood that a candidate Substance that adversely affects nism by which drugs and/or growth factorS operate; diag the tissue sample will not be suitable for use in the mouth. nosing and monitoring cancer in a patient, gene therapy; and These methods of the invention may thus be defined as the production of biologically active products, to name but methods for testing the oral toxicity or biocompatibility of a a few. For transplantation or implantation in Vivo, either the compound. oral tissues from the culture or the entire three-dimensional In an exemplary embodiment, the invention further pro culture could be implanted, depending upon the need. Three vides a dental test kit comprising, in Suitable container dimensional-tissue culture implants may, according to the means, a PLA, PGA or PLGA matrix in combination with a invention, be used to replace or augment existing tissue, to regenerated three dimensional dental pulp tissue Sample. introduce new or altered tissue, or to join together biological 35 Such a kit would be useful in methods for testing the tissueS or Structures. Suitability of a candidate Substance or compound for use in In yet another application, the three-dimensional culture dental practices Such as tooth filling or root canal proce System may be used as a "bioreactor” to produce cellular dures. products in large quantities, including products of exog Thus, the cultured oral tissue cells or regenerated tissue enous genes transferred into the cultured cells. For example, 40 Sample of the invention may be used in the preparation of a a cell which naturally produces large quantities of a particu medicament for use in reconstructing an oral tissue or for lar biological product (e.g., a growth factor, regulatory replacing lost or damaged oral tissue of an animal. In one factor, peptide hormone, antibody, etc.), or a host cell aspect of the invention, the medicament is intended for use genetically engineered to produce a foreign gene product, in reconstructing an oral tissue or for replacing lost or could be clonally expanded using the three-dimensional 45 damaged oral tissue of an animal at a site compatible with, culture system in vitro. If the transformed cell excretes the but distinct from, the oral tissue site from which the original gene product into the nutrient medium, the product may be Starting cells were obtained. In a further aspect of the readily isolated from the Spent or conditioned medium using invention, the medicament is intended for use in reconstruct Standard Separation techniques well known to those of Skill ing an oral tissue or for replacing lost or damaged oral tissue in the art. A "bioreactor' could be devised which would take 50 of an animal at the Same site from which the Starting cells advantage of the continuous flow method for feeding the were obtained. three-dimensional cultures in vitro. Essentially, as fresh In a particular embodiment of the invention, the cultured media is passed through the three-dimensional culture, the oral tissue cells or regenerated tissue Sample of the medi gene product will be washed out of the culture along with the cament are Separated from the matrix prior to application to cells released from the culture. The gene product could be 55 an animal. In a further embodiment of the invention, the isolated (e.g., by HPLC column chromatography, cultured oral tissue cells or regenerated tissue Sample of the electrophoresis, etc.) from the outflow of spent or condi medicament are applied to the animal in combination with tioned media. the matrix. Other uses are not excluded, Such as aiding in the diag In another embodiment of the invention, the cultured oral nosis and treatment of malignancies and diseases. For 60 tissue cells or regenerated tissue Sample of the medicament example, a biopsy of oral tissue may be taken from a patient are applied to the oral tissue site to be reconstructed or Suspected of having a malignancy. If the biopsy cells are replaced in the presence of an exogenous factor that Stimu cultured in the three-dimensional System of the invention, lates the growth or proliferation of the cells or tissue in the malignant cells will be clonally expanded during prolifera animal. In a particular aspect of the invention, the exogenous tion of the culture. This will increase the chances of detect 65 factor is produced by a cell that is also applied to the oral ing a malignancy and, therefore, increase the accuracy of the tissue site of the animal. The exogenous factor-producing diagnosis. Moreover, the patient's culture could be used in cell may be a recombinant cell engineered to produce Said 5,885,829 21 22 factor, or a cultured oral tissue cell of the cultured cells or preparation of a test kit for testing the Suitability of a regenerated tissue of the medicament, the cell being pro candidate Substance for use in the oral cavity. Vided with an exogenous gene that expresses Said exogenous factor. BRIEF DESCRIPTION OF THE DRAWINGS In another embodiment of the invention, the oral tissue cells of the cultured cells or regenerated tissue of the The following drawings form part of the present Specifi medicament are provided with an exogenous gene that cation and are included to further demonstrate certain expresses an exogenous factor in Said cells prior to appli aspects of the present invention. The invention may be better cation of the medicament to Said animal. In one aspect, the understood by reference to one or more of these drawings in oral tissue cells of the cultured cells or regenerated tissue of combination with the detailed description of Specific the medicament are provided with Said exogenous gene by embodiments presented herein. providing Said exogenous gene to the viable Starting cells FIG. 1. Photomicrograph of polyglycolic acid fiber-based before culturing the cells on the matrix, towards the begin mesh utilized as Synthetic extracellular matrix. A size bar is ning of the in vitro matrix culture process, or towards the end shown on the photomicrograph. of the in vitro matrix culture process. In certain aspects of the invention, the exogenous gene 15 FIG. 2. Photomicrograph of pulp-derived fibroblasts expresses an exogenous factor that Stimulates the growth or Seeded on polymer matrix. Cell-polymer constructs were proliferation of the oral tissue cells of the tissue Sample Visualized using a Scanning electron , and size when the medicament is applied to the animal. In other bars are shown on the photomicrographs. aspects, the exogenous gene expresses an angiogenic factor FIG. 3. Photomicrograph of pulp-derived fibroblast filling that stimulates the growth or proliferation of blood vessels the interstices between polymer . Cell-polymer con in or around the oral tissue cells of the tissue Sample when Structs were Visualized using a Scanning electron the medicament is applied to the animal. In yet other microScope, and size bars are shown on the photomicro embodiments of the invention, the exogenous gene graphs. expresses an exogenous therapeutic factor that is released by FIG. 4. Change in the cell density of cell-polymer con the cells of the tissue following application of the medica 25 Structs over time in culture. Values represent the mean ment to the animal. calculated from 2-4 Samples at each time point. In a further embodiment of the invention, the medicament FIG. 5A. The erosion kinetics (mass loss) of devices was is intended for use in filling a root canal. In an additional regulated by the polymer from which they were fabricated. embodiment, the medicament is intended for use in filling All masses were normalized to the initial device mass. the root canal of an existing tooth using cultured oral tissue Values represent the mean and Standard deviation calculated cells or a regenerated tissue Sample derived from an from 4 Samples at each time point. extracted tooth. In a further embodiment of the invention, FIG. 5B. The molecular weight (MW: o) of devices the medicament is intended for use in filling the root canal fabricated from 50/50 PLGA rapidly decreased in vitro. of an existing tooth using cultured oral tissue cells or a Glycolic acid (), D-lactic acid (O), and L-lactic acid (A) regenerated tissue Sample derived from a distinct, healthy 35 were released from the devices once the molecular weight molar or wisdom tooth. In yet another embodiment of the reached a low value. Values for the monomer release were invention, the medicament is intended for use in filling the normalized to the monomer mass initially present in the root canal of an existing tooth using cultured oral tissue cells device. The maximum monomer that could be released per or a regenerated tissue Sample derived from the same tooth. device was calculated using the measured initial device The cultured oral tissue cells or regenerated tissue sample 40 of the present invention may be used in combination with a mass. Values for the polymer molecular weight were nor Selected factor in the preparation of a medicament for use in malized to the initial molecular weight. delivering a Selected factor to an animal by administering FIG. 6. Representative strain diagrams of tubes formed the medicament to an oral tissue site of an animal. In one from the PGA mesh after Spraying with a Solution containing aspect of the invention, the Selected factor is produced by a 45 1% (O), 5% (), 10% (o), and 15% (O) PLLA. Devices cell and Said cell is included within Said medicament. In were subjected to a compressive force of 200 mN applied in another aspect of the invention, the Selected factor is pro a direction perpendicular to the axis of the device lumen duced by a recombinant cell engineered to produce Said Starting at 0 minutes. The force was removed at 10 minutes, factor and Said recombinant cell is included within Said and the change in the diameter of the tube (parallel to the medicament. 50 direction of force application) was monitored both during Alternatively, the Selected factor may be produced by an and after the time of force application, and normalized to the oral tissue cell of the cultured cells or regenerated tissue of initial diameter. the medicament, Said oral tissue cell being provided with an FIG. 7A. Representative strain diagrams of PGA tubes exogenous gene that expresses said Selected factor. In certain sprayed for various times (10 sec., D; 20 sec., O; 30 sec., ; aspects, the Selected factor is an orally therapeutic factor that 55 60 sec, o) with a 5% PLLA solution and subjected to a provides therapeutic benefit upon administration of the compressive force of 200 mN starting at 0 minutes. The medicament to an oral tissue site of an animal. In a particular force was removed at 10 minutes. The force application and aspect of the invention, the Selected factor is a Systemically the change in the diameter of the tube (normalized to the active therapeutic factor that provides Systemic therapeutic initial diameter) were monitored, as described in FIG. 6, benefit upon administration of the medicament to an oral 60 both during and after the time of force application. tissue site of an animal and Subsequent uptake into the FIG.7B. Devices sprayed with a 5% PLLA solution for 30 Systemic circulation of the animal. In a further aspect of the Seconds and tested dry (Control; o) or after pre-wetting for invention, the Selected factor is Subsequently removed from 24 hr in a saline solution (Pre-wet; O). The compressional Said animal by removing the medicament from the oral force was again 200 mN. tissue site of the animal. 65 FIG. 8. The degradation of devices bonded by spraying In an additional aspect of the invention, the cultured oral with PLLA or PLGA (5% solution; spraying time=30 sec.), tissue cells or regenerated tissue Sample is used in the as measured by quantitating the change in device mass over 5,885,829 23 24 time. Devices were incubated at 37 C. under static condi This invention provides the necessary methods for use in tions in buffered Saline, and removed at various times for creating new oral tissues, Such as dental pulp, using a tissue analysis. Values in represent the mean and Standard devia engineering approach. In the tissue-engineering of the tion calculated from 3 samples. PLLA, ; PLGA, O. invention, the cells of interest are isolated from a tissue, multiplied in culture, and Subsequently induced to form a FIG. 9. Release of ''I-labeled EGF from polymer micro new three-dimensional tissue. The invention particularly spheres. Results were normalized to the “I-labeled EGF concerns the use of Synthetic extracellular matrices in the incorporated into microSpheres. Values represent the mean culture and engineering of new oral tissues. and Standard deviation calculated from quadruplicate mea Current cell culture techniques, Such as those used in the SurementS. regeneration of Skin and/or oral mucosa, are not Suitable for FIG. 10A and FIG. 10B. FIG. 10A. Biological activity of use in connection with the oral tissues embodied in this EGF released from microspheres and control EGF. Quanti invention Since the existing techniques produce epithelia tation of the number of cells with labeled nuclei following which require an appropriate connective tissue bed in vivo H-thymidine autoradiography, and thus in S phase of the for Successful grafting. The techniques described herein cell cycle. Cells were cultured in medium containing various produce tissueS eX Vivo which will repair and regenerate concentrations of EGF which had not been incorporated into 15 microspheres (Soluble EGF), or in medium containing 10 Specific oral connective tissues in Vivo. ng/ml of EGF released from microspheres (Microspheres). While not desiring to be tied to any particular mechanism FIG. 10B. The change in the number of cells present in by which the invention works, a number of factors inherent culture dishes from day 1 to day 4 in medium containing in the three-dimensional culture System may contribute to its various concentrations of EGF which was not incorporated Success. Firstly, the three-dimensional framework provides a into microspheres (Soluble EGF), or in medium containing greater Surface area for protein deposition, and 10 ng/ml of EGF released from microspheres consequently, for the adherence of cells. Secondly, because (Microspheres). Values represent the mean and Standard of the three-dimensionality of the framework, cells continue error of the mean calculated from the results of 3 studies to actively grow, in contrast to cells in monolayer cultures, 25 which grow to confluence, exhibit contact inhibition, and which were all done in quadruplicate. The change in cell cease to grow and divide. The elaboration of growth and number was calculated relative to the cell number at day 1. regulatory factors by replicating cells may be partially FIG. 11A and FIG. 11B. Growth of fibroblasts over time responsible for Stimulating proliferation and regulating dif on matrices. FIG. 11 A. Growth of fibroblasts over time on ferentiation of cells in culture. Thirdly, the three PGA constructs. FIG. 11B. Growth of fibroblasts over time dimensional framework allows for a spatial distribution of within a collagen gel. Values represent the mean and Stan cellular elements which is more analogous to that found in dard deviation calculated from 3-4 Samples at each time the counterpart tissue in Vivo. Fourthly, the increase in point. potential Volume for cell growth in the three-dimensional FIG. 12. Proliferation of fibroblasts over time on coating System may allow the establishment of localized microen of adsorbed collagen. 35 vironments conducive to cellular maturation. Fifthly, the three-dimensional framework maximizes cell-cell interac DESCRIPTION OF ILLUSTRATIVE tions by allowing greater potential for movement of migra EMBODIMENTS tory cells, and for the establishment of communications In many situations, damage to oral tissues cannot be between the various types of cells in the adherent layer. effectively repaired. This leads to alternations in the form 40 Lastly, it has been recognized that maintenance of a differ and function of these tissues. Such alterations may give rise entiated cellular phenotype requires not only growth/ to deformities of the face, jaws and teeth which may be differentiation factors but also the appropriate cellular inter disfiguring as well as disabling. LOSS of teeth leads to actions. The present invention effectively recreates the tissue collapse of the dental arch and malpositioning of remaining microenvironment. teeth. Malpositioning may increase Susceptibility to diseases 45 Various issues are addressed in the oral tissue engineering Such as caries, gingivitis, and periodontitis which in turn can of the present invention. First, the appropriate cell types are lead to additional tooth loss. Tooth loss leads to diminished described; cells can be isolated from a variety of different mastication, and eventually to a diminution in the size of the tissue Sources, and expanded in culture. Second, Suitable jawbones rendering prosthetic reconstruction more difficult. matrices are described, for use in growing tissue ex vivo for It will be understood that oral tissue damage is painful to the 50 later Separation and use, and/or for use in transplanting the individual and costly to Society. matrix and tissue into the body to stimulate further cells to Injury or infection of adult dental pulp often necessitates grow into the matrix from the Surrounding tissue. Third, root canal therapy. This terminates dentin formation and methods for using the tissues and matrix materials in an Subsequent tooth maturation. Unfortunately, the Synthetic integrated manner are described in order to create a tissue materials currently utilized to replace lost tooth Structure are 55 replacement with the appropriate Structure and function. not capable of completely replacing the function of the lost Screening assays are also described. tissue, and often fail over time. The engineering of dental A. Cells and Tissues pulp and other oral tissues from cultured cells would thus In the present invention, viable cells are obtained from a represent a marked Step forward in dentistry and oral medi Specific oral tissue and are cultured in vitro. In general, in cine. 60 tissue engineering with other tissues, a first question is often If effective oral tissue engineering techniques could be whether to transplant cells to a desired Site, or to provide an developed they would provide an effective alternative to environment inductive or conductive to the formation of the many of the present inadequate treatment Strategies. These desired tissue from cells already present in the host tissue. include many dental procedures, areas related to periodontal The present invention provides methods that are suitable for disease and, also, more Substantial procedures, Such as the 65 achieving both goals-as it provides tissue transplantation repair of resectioned tissue, for example upon removal of itself and, if desired, a matrix Structure to induce further cancerous growths and tumors. tissue formation. 5,885,829 25 26 1. Sources of Cells cells, Schwann cells, endothelial cells, and undifferentiated In certain embodiments of the present invention, the cells mesenchymal cells. Cells involved in the immune response, for use in tissue regeneration and transplantation will be Such as macrophages, mast cells, antigen processing cells obtained from an oral tissue site of the animal or patient to (dendritic cells), and plasma cells can also be found in the be treated. These are “autologous cells' that give rise to pulp during periods of inflammation. “autologous tissues”. However, the use of cells from another Odontoblasts are terminally differentiated, polarized pull animal of the same species (or person) is certainly not pal cells derived from the cranial neural crest, which are excluded. These would be “allogeneic cells and tissues”. found in a peripheral layer closely associated with the Further, cells from tissueS of other animals, "Xenogeneic cells and tissues' could also be used. predentin. The major function of odontoblasts is the Syn The use of autologous cells eliminates concern over cell thesis and Secretion of the fibers and extracellular matrix rejection, avoids the issue of immunosuppreSSants, and is (ECM) of the predentin and biomineralization of the dentin. attractive in a variety of Scenarios. Although two procedures The cell bodies form an irregularly columnar, epithelial on the animal or patient are required, this does not cause a like layer on the inner aspect of the dentin. The proximal particular disadvantage in dentistry and oral medicine as the Surface of the cell body is adjacent to pulp cells, and the tissue sites are easily accessible. The time required to 15 distal extremity is tapered and embedded in predentin, an expand the cells into a tissue Sample, which creates a unmineralized layer of dentin-like material. time-lapse before treatment can be completed, is also not of The major protein produced by the odontoblast is type I any particular concern in dentistry and oral medicine as any collagen and is Secreted into the extracellular space at the acute disease or injury requiring immediate therapy can predentin interface. Non-collagenous components of the likely be treated with a temporary measure in the intervening extracellular matrix of predentin and dentin, including time period. proteoglycans, glycosaminoglycans, phosphoproteins, and AS allogeneic tissue is widely used in humans in whole Y-carboxyglutamate-containing proteins, are also Synthe organ transplantation, it is also a viable Source for cells for sized and Secreted by Odontoblasts. use in the present invention. The tissue could be immedi The functioning odontoblasts continue to produce preden ately available for use in large quantities in a variety of 25 tin throughout the life of the tooth. Odontoblasts retain the procedures. Immunosuppressive drugs are available if ability to upregulate protein Synthetic activity in response to required to prevent tissue rejection. The use of Xenogeneic trauma after aging. cells is not excluded from the Scope of this invention, The remainder of the pulp consists of a “stromal' tissue although the present invention is advantageous in that it is containing nerves, blood vessels, and lymphatics. The Stro particularly Suitable for use with autologous cells. mal tissue is composed of cells and extracellular material. 2. Oral Tissues There appears to be only one type of cell, resembling Each tooth consists of a crown and either Single or mesenchyme but capable of producing extracellular multiple roots. By definition, the anatomical crown is cov material, including collagen. Thus, the cell can equally be ered by the highly calcified layer of enamel. The remainder termed a fibroblast, or simply a pulpal cell. of the crown is composed of another calcified tissue, the 35 Fibroblasts are the most numerous cells found in the dentin, which contains a central chamber filled with the dental pulp. They are Stellate-shaped cells with long cyto living tissue of the tooth, the pulp. The dentin and pulp also plasmic extensions that contact adjacent fibroblasts or odon constitute the tissueS of the root. However, the outer Surface toblasts through gap-junctional processes. Fibroblasts Syn of the root is covered by yet another calcified tissue termed thesize and Secrete type I and type III collagen, and other cementum. The enamel and cementum meet around the 40 ECM components of the pulp, including proteoglycans and neck, or cervical margin, of the tooth and form the cemento glycosaminoglycans. enamel junction. This junction is not normally Seen on the Collagen is the most abundant connective tissue protein portion of the tooth exposed to the oral cavity because the and occurs in Several Specific isotopes, types I through XII. epithelial tissue covering the alveolar process, the gingiva, Each is recognized as a specific genetic product differing in extends for a short distance onto the crown. 45 amino acid and polypeptide composition. In the pulp, type I The dental pulp consists of loose connective tissue and type III are the most abundant, with other types, Such as derived from ectomesenchymal cells and is confined within IV and V, as minor constituents. the pulp chamber and root canals of the tooth. The pulp Fibroblasts or undifferentiated mesenchymal cells also contains cells that provide odontogenic, nutritive, Sensory, have an important role in wound healing mechanisms in the and defensive functions to the mature pulp and allows for 50 pulp. The fibroblasts of the cell-rich Zone are thought to preservation of Vitality during normal homeostatic mainte differentiate into odontoblasts after the right stimulus-for nance and during wound repair after injury. example, growth factor, a bone morphogenic protein (BMP), The mature dental pulp can be divided into two compart cytokine, or inflammatory mediator, released during wound ments: The Odontogenic Zone and the pulp proper. The ing from the exposed predentin or dentin, or inflammatory odontogenic Zone includes the odontoblasts, which are the 55 cells that have migrated to the wound Site. cells responsible for the production of predentin and dentin, There are many other cells found in a vital dental pulp. the cell-free Zone, the cell-rich Zone, and the parietal plexus Perivascular cells are found in the dental pulp closely of nerves. The pulp proper includes the majority of the asSociated with the vasculature. These cells have been remaining area of the pulp and consists primarily of fibro reported to be important in wound-healing mechanisms blasts and extracellular matrix, blood vessels, and nerves. AS 60 asSociated with pulpal repair mechanisms. Perivascular cells the pulp ages, the Volume decreases with a corresponding have also been shown to proliferate in response to an increase in dentin thickness. Numerous Studies have dem iatrogenic exposure of the dental pulp, and are thought to onstrated that the dental pulp has an inherent capacity to possibly provide replacement cells for the odontoblast layer respond to wounding in the absence of other inflammatory in wounds where the cell-rich layer has been destroyed. insults. 65 Endothelial cells line the lumen of the pulpal blood The most predominant cell type in the dental pulp is the vessels and contribute to the basal lamina by producing type fibroblast, but the pulp also contains odontoblasts, blood IV collagen, an afibrillar collagen. They have been shown to 5,885,829 27 28 proliferate after a pulp exposure in an attempt to neovascu HemidesmoSomes are responsible for the attachment of larize the wounded area during the process of wound heal the gingiva as a continuous cuff around the tooth. This ing. attached cuff fulfills the essential function of sealing off the Class II antigen processing cells have been demonstrated access route from the mouth to the periodontal tissues. by immunohistochemical methods in both the normal and Breakdown of this Seal results in gingival infections inflamed pulp. Other vascular-derived cells found in the pulp (gingivitis) and invasion by microorganisms, leading to during an inflammatory condition include mast cells, B- and periodontal disease. T-lymphocytes, polymorphonuclear neutrophils, and mac By understanding oral tissue anatomy, as outlined above, rophages. These blood cells are of paramount importance in and in light of the present disclosure, one of ordinary skill fighting infection in the pulp because of the Substances they in the art will readily be able to Select an appropriate tissue contain: histamine, Serotonin, cytokines, growth factors, and Source for use in the tissue regeneration methods disclosed other cellular mediators. Schwann cells can also be found, herein. which cells envelope nerve processes with a myelin sheath. 3. Viable Cells Dentin forms the bulk of the tooth. It is lined on its outer The methods of the present invention generally require aspect by enamel on the crown and by cementum on the root. 15 the isolation of compositions comprising viable cells from The youngest layer of dentin formed at any particular time oral tissues. In this context, the term “viable cells' means is adjacent to the junction between the Odontoblast cell body that the cells are capable of proliferating and, preferably, and its major process. This layer of young dentin is essen regenerating to form an oral tissue when Subjected to the tially unmineralized, and ends abruptly in contact with the methods described herein. The choice of appropriate viable mature dentin. The predentin is an irregular meshwork of cells will be readily determinable to one of skill in the art. collagenous fibrils that tend to be thin closer to the cell body For example, when the intention is to culture and regenerate and Somewhat larger in diameter near the mature dentin. dental pulp tissue, one will obtain a composition comprising Mature dentin is associated with glycoproteins, an viable cells that are capable of producing Such dental pulp increased diameter of the collagenous fibrils, and a Sudden tissue or gingival Submucosal fibroblasts. This is exempli and dramatic mineralization related to these fibrils. The layer 25 fied in the present invention by obtaining a tissue Sample that of mature dentin is very thick in functional teeth. comprises dental pulp-derived fibroblasts. However, this is The outer surface of the root is covered by a relatively thin just one exemplary embodiment of the present invention. layer of a bone-like mineralized tissue called cementum. It will be understood by those of skill in the art that the Cementum consists of a matrix of calcified collagenous tissue regeneration methods of this invention are widely fibrile, glycoproteins, and mucopolysaccharides. The Outer applicable to the culture of Several oral tissues. One may most layer of cementum is an uncalcified precementum mention, by way of example only, dentin, periodontium, produced by the discontinuous layer of irregularly shaped gingival Submucosa, oral Submucosa and tongue tissues. cementoblasts. Naturally, one would choose a starting composition from an The firm epithelium from the mucogingival junction (line) appropriate location in order to ensure that the composition to the teeth is the gingiva. It extends around each tooth, and 35 contains cells capable of regenerating into the desired tissue meets the regular oral mucosa at a less well-defined inner Structure. mucogingival junction. The Source of dental pulp for use in tissue regeneration Gingiva is a Stratified Squamous epithelium with deep may be from a damaged tooth that Still contains certain papillary projections from the underlying lamina propria. viable cells or from another, healthy tooth, distinct from the The latter contains a rich capillary network that is conse 40 tooth that requires the restorative procedure. The Odonto quently brought relatively close to the Surface. The epithe blasts of the dental pulp are also Suitable for use in gener lium is firmly attached to the lamina propria by hemides ating dentin. mosomes related to the basal lamina and collagenous The Source of cells for the regeneration of the periodon anchoring fibrils that originate from the hemidesmoSomes tium is that portion of the tissueS which remains attached to and form loops in the lamina propria. Collagenous fibrils 45 a freshly extracted tooth. These tissue fragments containing from the lamina propria itself pass through these loops, thus viable cells are Scraped from the tooth root Surface using a contributing to the firm attachment between the two tissues. Sharp instrument Such as a curette or Scalpel blade. The basal layer of the epithelium is cuboidal and shows For the gingiva and oral mucosa, Superficial Split numerous mitotic figures, which provide for the renewal of thickness biopsies of these tissues are obtained by routine this epithelium. Toward the surface of the epithelium, the 50 clinical procedures. The tissues are suspended in DMEM, cells flatten to form a compact layer in which the cells are 10% fetal bovine Serum, Supplemented with agents Such as, partially filled with keratin but the nucleus and some e.g., penicillin, Streptomycin and neomycin, and minced to organelles Survive. Thus, the dozen or So cell layers at the pieces approximately 1–2 mm, and placed into tissue Surface are keratinized but not dead. culture vessels. This is best done within 24 hours of biopsy. This epithelium differs from that of the skin, where the 55 When sufficient numbers of cells have migrated from the resulting layer of dead, cornified cells forms the dry Surface cultured tissue pieces (explants) and proliferated, the cul of the skin. The gingival generative process, which is called tures are harvested diluted into more vessels and/or cryo parakeratosis, is thus different to the orthokeratosis of the preserved. Cells may also be obtained by mincing the skin. Gingiva is a strong resilient epithelium that turns over tissues, and then digesting the pieces with enzymes Such as rapidly enough to withstand and heal the trauma and abra 60 collagenase. Sion caused by chewing. In order to regenerate an oral tissue in accordance with the The lamina propria of the gingiva is firmly attached to the present invention, one may obtain a composition consisting periosteum of the alveolar bone except as it approaches to only of, or predominantly of, healthy, viable cells. Using the within 1 or 2 mm of the crown Surface. This narrow band of regeneration of dental pulp as an example, Such a compo gingiva Surrounding each tooth is called the “free gingiva 65 Sition could be obtained by isolating dental pulp from a as opposed to the rest of the gingiva, which is “attached” to healthy tooth of an animal, Such as a healthy molar or the alveolar bone. wisdom tooth. The viable autologous cells would then be 5,885,829 29 30 regenerated into a tissue Sample that could be reapplied to a dishes, Vials and other receptacles are not “matrices' in the diseased or damaged tooth of the Same animal. context of the present invention, although it is possible that However, it is important to note that the above technique the materials from which they are formed may be adapted is not the only embodiment of the present invention. In fact, for this purpose by design and/or combination with other the invention may be used to regenerate healthy oral tissues, matrix elements. Such as dental pulp, by obtaining a less than completely The materials utilized to fabricate a matrix for use in the healthy oral tissue and regenerating the viable cells from present invention can generally be categorized into three Said tissue by culture on a matrix, as described herein. These types: naturally derived materials, including extracellular aspects of the invention are based, in part, on the fact that matrix (ECM) molecules, Such as collagens and hyaluronic cells are routinely cultured from oral tissues which are acid, and polysaccharides, Such as alginate, Synthetic coated with large numbers of microbes Such as yeast and materials, including any one of a variety of polymers, and bacteria. Since these microbes proliferate in the cell culture relatively new materials that incorporate Specific cell rec media, antibiotics are routinely added at up to 3 times the ognition signals found in ECM molecules. normal concentrations of antibiotics and antimycotics. 1. Naturally-Derived Matrices Another embodiment of the present invention utilizes 15 Any one of a variety of naturally-derived matrix-like viable cells isolated from oral or gingival Submucosa to materials may be used to provide a framework for tissue regenerate other tissueS Such as dental pulp, dentin, growth in accordance with the present invention. Where the cementum, periodontal ligament and bone. Such cells propa matrix, or Substantial portions thereof, will later be reapplied gated in vitro, Seeded onto Scaffold and developed in vitro, to an oral tissue site in the body, one will generally prefer to would be constitutively capable, genetically engineered or use a matrix that is derived from a biological tissue that is induced by the addition of Specific protein factors to produce compatible with the tissue to which it will be readministered. the Specific tissue of interest when implanted in Vivo. Such biocompatibility requires that the matrix does not In general terms, following isolation of an oral tissue cause any significant adverse or untoward reactions when Sample that contains the desired viable cells, the composi administered to the animal. By using a biocompatible matrix tion would first be cultured in vitro in order to expand the 25 Significant immune responses and inflammatory reactions population of viable cells. Although not required as part of will be avoided. the regenerative methods of the invention, this pre-culture A large number of naturally-derived matrix-like materials Step is generally recommended in order to provide a larger are available that may be used in tissue regeneration in population of Starting cells. accordance with this invention, including those matrices A wide variety of Standard cell culture techniques are fabricated from human, animal or plant tissue. Potential available that may be used in this aspect of the invention. advantages of these types of materials are their biocompat Virtually any culture medium and technique may be used, So ibility and their biological activity. AS many of these mol long as the chosen method results in maintenance, or pref ecules are found within tissues, they may not induce any erably proliferation, of the viable cells that one wishes to foreign body reactions and are presumably receptive to the regenerate. Such techniques will be known to the ordinary 35 cell-mediated remodeling that occurs during tissue repair skilled artisans. Further, the texts of Freshney (1994) and and regeneration (Murphy et al., 1990; Yannas et al., 1989). Janda et al., (1991) are fully incorporated herein by refer The primary, Secondary and tertiary Structure of a variety ence to even further describe animal cell culture techniques. of ECM molecules have been defined, as have specific Although a full discussion of Standard cell culture tech cellular molecules involved in ECM recognition. Cells con niques and media is not believed to be necessary as part of 40 tain transmembrane receptors that specifically bind defined the present disclosure, one may mention, by way of amino acid sequences present in ECM molecules (Hynes, example, the use of higher than usual antibiotics in the cell 1987). The binding of cellular receptors to ECM molecules culture procedures. The use of up to about 2 or 3 times Starts a Sequence of events that can alter both cell growth and higher than the normal concentrations of antibiotics and tissue-specific gene expression (Schwartz and Ingber, 1994). antimycotics is believed to be particularly effective. In using 45 A variety of growth factors are also known to associate with a tissue Sample from an infected tooth, e.g., during a root the ECM in tissues. ECM molecules may serve as a reposi canal procedure, increasing the amounts of antimicrobials tory of these factors for cells adherent to the ECM (Folkman will generally be preferred. and Klagsbrun, 1987; Reid, 1990; Stoker et al., 1990). One The use of antimicrobial agents that are readily available may precisely regulate where transplanted or induced cells and commonly used, Such as penicillin, Streptomycin, neo 50 adhere to the matrix, and their exposure to specific growth mycin and erythromycin, is generally preferred, although factors by utilizing specific ECM molecules to fabricate a any one or a combination of antimicrobials may be matrix. This may allow the gene expression of cells present employed, as desired. The penicillins are known to be in the matrix to be tightly regulated. effective against certain species of Gram-positive cocci, Type I collagen, the most prevalent ECM molecule in the Gram-negative cocci, Gram-positive bacilli and Gram 55 body, is readily isolated from animal tissues and has been negative bacili. extensively utilized to fabricate cell delivery devices (Green B. Matrices et al., 1979; Yannas et al., 1981; Bell et al., 1981; Stern et After obtaining the cells, culture and tissue regeneration al., 1990; Cavallaro et al., 1994). This material can be generally involves the use of a Structural matrix. The matrix processed into a wide variety of Structures for use in the acts as a Scaffold for the cells to guide the process of tissue 60 present invention, e.g., films, Sponges and fibers (Green et formation. Although the majority of mammalian cell types al., 1979; Yannas et al., 1981; Bell et al., 1981; Stern et al., are anchorage dependent, and will die if not provided an 1990; Cavallaro et al., 1994). The structure and resultant adhesion Substrate, the matrices of the present invention are mechanical properties of collagen-based Scaffolds can be not simply an adhesive substrate. Petri dishes and other regulated by the proceSS utilized to extract the collagen from non-matrix Structures are generally used in cell culture, 65 tissues (Cavallaro et al., 1994), and by various crosslinking whereby cell monolayerS result, however, this does not lead processes. Collagen molecules may be crosslinked physi to tissue regeneration. It will be understood that Simple cally by dehydrothermal (Koide et al., 1993) or UV radiation 5,885,829 31 32 treatments, or chemically by using various chemical agents Synthetic polymers can be processed with various techniques (Cavallaro et al., 1994; Koide et al., 1993; DeLustro et al., and Supplied consistently in large quantities. The mechanical 1990). However, the inflammatory response to these mate and physical properties of Synthetic polymers can be readily rials and their erosion rate are dependent on the Specific adjusted through variation of molecular structures So as to cross-linking agent that is utilized (Cavallaro et al., 1994, fulfill their functions without the use of either fillers or Anselme, 1992; Koide et al., 1993). additives. Table 1 outlines different structural factors of Suitable collagen matrices are described, for example, in polymers that can be used to adjust a variety of critical U.S. Pat. Nos. 4,347,234; 4,390,519; 4,394,370; 4,409,332; properties. 4,538,603; 4,585,797; 4,703,108; 4.837,285; 4,975,527; 5,081,106; 5,128,136; 5,162,430; 5,197,977 and 5,206,028; each incorporated herein by reference. Although not previ TABLE 1. ously proposed for use regenerating oral tissueS eX Vivo, the Structural Variables used to Control Biodegradable Polymer Properties biocompatibility of collagen matrices is thus well known in the art. If desired, therefore, a collagen-tissue preparation Variables Effects Examples could also be applied to a tissue site of an animal. Miner 15 Incorporation of both May reduce/eliminate Non-immunologic PGA alized collagen, as disclosed in U.S. Pat. No. 5,231,169, natural and/or non immunologic response and PLA (vs. collagens) incorporated herein by reference, may also be used in the natural monomers often found in present invention. naturally-derived Type I collagen may also be combined with glycosami polymers Incorporation of labile Control kinetics of Hydrolyzable ester bond noglycans to form gels which mimic native dermal tissue groups in polymer biodegradation in PGA (Yannas et al., 1981; Stern et al., 1990; Heimbach et al., chain 1988). A variety of other ECM molecules, including laminin Incorporation of Control chemical and Hydrophilic, hydro (Dixit, 1994; Guenard et al., 1992), have been utilized as cell functional groups in physical properties of phobic and amphiphilic delivery matrices, and any Such matrix may be used in the side chains polymers polyphosphazenes Incorporation of Control physical and Semi-crystalline L-PLA context of the present invention. chiral centers in mechanical properties and amorphous D.L-PLA Polysaccharides may also be used as matrices in accor 25 polymer chains of polymer dance with this invention. Alginate, a polysaccharide iso Possibility of utilizing Control properties of Glycolic and lactic acids lated from seaweed, is used as a cell delivery vehicle. Water multiple monomers polymers PLGA Use of natural com Biocompatible break Lactic acid in PLA Soluble Sodium alginate readily binds calcium, forming an pounds as monomers down products insoluble calcium alginate hydrocolloid (Sutherland, 1991). Use of different Control physical and Branched polymers ex These gentle gelling conditions have made alginate a popu polymer architectures mechanical properties hibit lower viscosity than lar material to encapsulate cells for transplantation (Lim and of polymers linear ones Sun, 1980; O'Shea et al., 1984; Ricordiet al., 1988; Sullivan et al., 1991; Lacy et al., 1991; Levesque et al., 1992; Adapted from Wong and Mooney, 1997. Soon-Shiong et al., 1994; Dixit, 1994; Kasai et al., 1994), A variety of Synthetic biodegradable polymers can be and as an injectable cell delivery vehicle (Atala et al., 1994). 35 utilized to fabricate oral tissue engineering matrices. In The potential advantages of these natural materials have general, these materials are utilized as Structural elements in made them popular for fabricating tissue engineering the scaffold, to deliver the tissue, or to achieve both pur matrices, and they may certainly be used in the context of the poses. Poly(glycolic acid) (PGA), poly(lactic acid) (PLA) present invention. However, these materials also have a and poly(lactic acid)-poly(glycolic acid) (PLGA) polymers number of disadvantages. Many of these materials are 40 are commonly used Synthetic polymers in tissue engineer isolated from human or animal tissue, and are not available ing. These polymers are also extensively utilized in other in large quantities. They Suffer from large batch-to-batch biomedical applications Such as drug delivery and are FDA variations, and are typically expensive. Additionally, these approved for a variety of applications (Huang, 1989). materials exhibit a limited range of physical properties (e.g., A number of PGA, PLA and PLGA and other synthetic mechanical Strength, erosion times). These drawbacks led 45 polymer matrices are known in the art, and are further the present inventors to contemplate using Synthetic mate described herein, any one or more of which may be used in rials to fabricate matrices for use in many aspects of this the context of the present invention. By way of example invention. only, one may mention the PGA, PLA and PLGA formula 2. Synthetic Matrices tions disclosed in any one of U.S. Pat. Nos. 5,366,734; In certain aspects, Synthetic polymers are attractive Scaf 50 5,366,733; 5,366,508; 5,360,610; 5,350,580; 5,324,520; fold materials as they can be readily produced with a wide 5,324,519; 5,324,307; 5,320,624; 5,308,623; 5,288,496; range of reproducible properties and Structures. Polymer 5,281,419; 5,278,202; 5,278,201, 5,271,961; 5,268,178; matrices also provide mechanical Support against compres 5,250,584; 5,227,157; 5,192,741; 5,185,152; 5,171,217; Sive and tensile forces, thus maintaining the shape and 5,143,730; 5,133,755; 5,108,755; 5,084,051; 5,080,665; integrity of the Scaffold in the aggressive environments of 55 5,077,049; 5,051,272; 5,011,692; 5,007,939; 5,004,602; the body. 4,961,707; 4,938,763, 4,916, 193; 4,898,734; 4,898,186; The morphology of the matrix can guide the Structure of 4.889,119, 4,844,854; 4,839,130; 4.818,542; 4,744,365; an engineered tissue (Vacanti et al., 1988), including the 4,741,337; 4,623,588; 4.578,384; 4,568,559; 4,563,489; size, shape and vascularization of the tissue (Mooney et al., 4539,981; 4,530,449; 4,384,975; 4,300,565; 4,279,249; 1994a, 1994b, 1995b, 1996a). The ideal matrix should also 60 4,243,775; 4,181,983; 4,166,800; 4,137,921 each incorpo elicit specific cellular functions and direct cell-cell interac rated herein by reference. tions. Tissue engineering matrices function as Synthetic Where a matrix is to be administered to an oral tissue site, extracellular matrices, and the proper design of these matri another reason for preferring a Synthetic material is that the ceS allows them to exhibit the required range of mechanical Surface properties of Synthetic materials can be easily and and biological functions. 65 reproducibly altered, as necessary. Plasma modification and Synthetic polymeric materials can usually be precisely grafting of relatively inert Substances, Such as polyethylene controlled in material properties and quality. Moreover, oxide or polyvinyl alcohol, can mask the chemistry of the 5,885,829 33 34 bulk matrix (Peppas and Langer, 1994). The specific struc polymer matrices that may be used in this invention. Further ture of adsorbed polymer coatings can be controlled by biodegradable matrices include polyanhydrides, varying the chemical Structure and molecular weight poly polyorthoesters, and poly(amino acids) (Peppas and Langer, dispersity of the coating polymer (Dan and Tirrell, 1993). 1994). Any such matrix may be utilized to fabricate a Molecular Self-assembly Strategies can also be used to define biodegradable polymer matrix with controlled properties for the protein and cellular interactions with material Surfaces use in this invention. Further biodegradable polymers that (Prime and Whitesides, 1991; Singhvi et al., 1994). produce non-toxic degradation products are listed in Table 3. 3. Biodegradable Matrices Matrices fabricated from biodegradable materials will TABLE 3 erode over time in the body to yield a completely natural Main Polymers Recognized as Biodegradable tissue. These matrices will not induce any chronic inflam matory responses, and cannot Serve as a long-term Site for Synthetic infection. Biodegradable polymers have been utilized to Polypeptides engineer tissues that will be structurally integrated with the Polydepsipeptides host tissue (Langer and Vacanti, 1993; Heimbach et al., 15 Nylon-2/nylon-6 copolyamides 1988; Hansbrough et al., 1992; Mooney et al., 1994a; 1994b; Aliphatic polyesters 1995a; 1995b; 1996a; Johnson et al., 1994, Dixit, 1994; Poly(glycolic acid) (PGA) and copolymers Kasai et al., 1994; Mooney and Vacanti, 1993). In addition, Poly(lactic acid) (PLA) and copolymer the use of synthetic, biodegradable matrices will often be Poly(alkylene succinates) advantageous as the degradation time of Such Synthetic Poly(hydroxybutyrate) (PHB) matrices can be designed to coincide with the formation of Poly(butylene diglycolate) a new tissue from the cultured cells. Poly(e-caprolactone) and copolymers While there are a variety of biodegradable polymers Polydihydropyrians (Gilding, 1981; Peppas and Langer, 1994), polymers com Polyphosphazenes posed of monomers naturally present in the body (e.g., lactic Poly(ortho ester) 25 Poly(cyano acrylates) acid, C.-amino acids) are preferred for use in certain aspects Natura of the invention. Polymers of lactic acid, glycolic acid, and copolymers of the two have been utilized to fabricate tissue Modified polysaccharides engineering matrices (Heimbach et al., 1988; Hansbrough et cellulose, starch, chitin al., 1992; Mooney et al., 1994a; 1994b; 1995a; 1995b; Modified proteins 1996a; Johnson et al., 1994; Mooney and Vacanti, 1993). collagen, fibrin These polymers are readily processed into a variety of Adapted from Wong and Mooney, 1997. configurations, including fibers (Frazza and Schmitt, 1971), porous sponges (Mooney et al., 1995a; Mooney and Vacanti, Although the preferred use of the biodegradable matrices 1993) and tubular structures (Mooney et al., 1995b). in the present invention is for administration to an oral tissue The regular structure of homopolymers of lactic and Site in conjunction with an oral tissue Sample, the biode glycolic acid results in a crystalline Structure (Gilding, 35 gradable matrices may also be used to regenerate an oral 1981). Copolymers containing significant quantities of both tissue Sample that is Subsequently to be isolated free from monomers are amorphous (Gilding, 1981). This polymer the matrix. The tissue Sample may then be readministered to family's widely varying mechanical and erosion properties an oral tissue site of an animal. (Table 2) results both from the varying crystallinity, and the It will of course be understood that biodegradable matri differing hydrophobicity of lactic and glycolic acid (Gilding, 40 ceS for use in the invention are not confined to being 1981). Regenerated oral tissues of this invention can thus be Synthetic matrices. A number of naturally-derived matrix delivered using matrices with a wide range of pre-defined like materials may be used that will eventually biodegrade in degradation times and mechanical properties, where the an in Vivo environment. Thus, in the context of the present matrices are fabricated from this family of polymers invention, the term biodegradable is not necessarily Synony (Mooney et al., 1995b). 45 mous with Synthetic matrices. 4. Non-Biodegradable Matrices TABLE 2 Although biodegradable matrices will have advantages in certain embodiments, they are by no means required for use Typical Yield Stress Values and Erosion Times for Polymers in practicing the present invention. Those of skill in the art of Lactic and Glycolic Acid 50 will understand that there are at least two Scenarios in which YIELD STRESS TIME FOR 50% non-degradable matrices are envisioned for use. First, a POLYMER (Kpsi)* EROSIONf non-degradable matrix may be used to regenerate an oral tissue Sample that is Subsequently to be isolated free from polyglycolic acid 11.2 4 weeks the matrix. The tissue Sample may then be readministered to 50/50 poly (D.L-lactic-co-glycolic 7.7 6 an oral tissue site of an animal. The function of the matrix acid) 55 85/15 poly (D.L-lactic-co-glycolic 6.3 2O in this context thus ceases once the tissue has been regen acid) erated and removed from the Structural matrix. poly (D.L-lactic acid) 6.6 35 In a Second embodiment, the regenerated oral tissue poly (L-lactic acid) 8.5 >56 Sample may remain in contact with the matrix and the matrix-tissue preparation may be administered to the Adapted from Wong and Mooney, 1997. 60 *Values represent the mean of 5 measurements obtained using Instron testing required oral tissue site of an animal. In clinical Situations with ASTM methods. Data is adapted from Medisorb (Cincinnati, OH) that currently employ the use of a permanent Synthetic plus data. material implant, the administration of an implant-tissue The time at which 72 of the polymer has eroded polymer mass = % initial combination would not, of course, be associated with any mass) following immersion in a buffered saline solution maintained at 37 C. disadvantages. However, it will also be appreciated that Such 65 non-degradable matrices will need to be biocompatible if Those of skill in the art will understand that the PLA, PGA they are to be administered to an animal. This is in contrast and PLGA polymers are just one example of biodegradable to the first situation described above, where the isolation of 5,885,829 35 36 the regenerated tissue from the matrix means that the cial proteins with a desired backbone structure and amino biocompatibility of the original matrix is generally irrel acid Side chains that promote cell adhesion (McGrath et al., eVant. 1992). These artificial proteins can be expressed in bacterial Calcium phosphate ceramics are non-biodegradable cells, isolated and purified, and utilized to form matrices or matrices that are extensively used in engineering bone tissue coat other Surfaces (Anderson et al., 1994). This approach (Ducheyne, 1988) and may be used in the present invention. offers tremendous control over both the properties (bulk and A suitable ceramic that may be used is described in U.S. Pat. Surface) of the material, and its ability to interact with cells. No. 4,596,574, incorporated herein by reference. Traditional Synthetic routes are also being used to develop Both hydroxyapatite and tricalcium phosphate, and mix biodegradable polymers that contain cell recognition pep tures of the two, may be utilized. These materials can be tides as side chains (Barrera et al., 1993). The advantages of coated over metal implants (Lemons, 1988), used with the Synthetic polymers, Such as polylactide, can be combined tissue implant as an additional bone inductive or conductive with the specific biological activity of ECM molecules with material (Ducheyne, 1988; Jarcho, 1981), or used as a cell this approach. A similar approach is the Synthesis of short delivery vehicle (Goshima et al., 1991). These materials amino acid chains containing a desired functional group that only release calcium and phosphate as breakdown products. 15 can be covalently bonded or adsorbed onto matrices fabri They display no local or Systemic toxicity, and become cated from other synthetic materials (Hubbell, 1995). Such directly bonded to adjacent bone tissue with no intervening biomimetic Synthetic polymers and cell-adhesion peptides fibrous capsule (Ducheyne, 1988). The erosion and are proposed for use as implants for tissue regeneration and mechanical properties of these materials are controlled by transplantation (Hubbell, 1995). These and any of the fore the Specific chemical composition and processing conditions going or other “Second generation' matrices may be used in (Lemons, 1988). the context of the present invention. Applications of hydroxyapatite, tricalcium phosphate, and Bone achieves its high mechanical moduli by combining mixtures thereof are currently limited by their brittle nature organic and inorganic materials, and this principle is being and generally poor mechanical properties (Jarcho, 1981). utilized to Synthesize new ceramic materials. Apatite crys However, this is not a drawback in the context of the present 25 tals are being Synthesized by nucleation and growth around invention. First, where the tissue is to isolated from the poly(amino acids) (Stupp and Ciegler, 1992). This results in matrix prior to use, this would not be a significant limitation. an intimate dispersion of the organic molecules within the Further, where both the matrix and the regenerated oral ceramic, and improves the mechanical properties of the tissue are to be administered, the presence of the natural ceramic (Stupp et al., 1993). This process may mimic the tissue will negate any drawbacks that the matrix may have, process of natural bone formation, and these materials show as the matrix will be a Supplement to the tissue, not the only promise in engineering bone tissue (Stuppet al., 1993). Such material to be relied upon. materials may also be used in aspects of this invention. Further non-biodegradable polymers include Semiperme C. Regeneration Methodology able polymerS Such as poly(acrylonitrile-co-Vinyl chloride) The regeneration methodology first involves cell Seeding (Emerich et al., 1992; Sagan et al., 1993; Guenard et al., 35 onto the three dimensional matrix. Cells can be seeded under 1992), polylysine (Lim and Sun, 1980; O'Shea et al., 1984; Static or Stirred conditions. Cells can be seeded in a one time Ricordi et al., 1988; Sullivan et al., 1991; Lacy et al., 1991; administration, continuously added, or added in batches. Levesque et al., 1992; Soon-Shiong et al., 1994), cellulose Any desired method and apparatus may be used to Seed the acetate (Yang et al., 1994) and polysulfone (Yang et al., cells onto the matrix. Exemplary methods are described 1994). Although generally intended for use in immobilized 40 herein in the detailed examples. In Stirred conditions, a cell cells, the use of Such polymers in the context of the present Suspension is mixed with Scaffolds and agitated to enhance invention is certainly not excluded. These polymerS may the number and frequency of cell contacts with the matrix to also be used with a variety of gels, including alginate and maximize cell adhesion to the matrix. polyphosphaZenes. As will be appreciated by those of skill in the art, 37 C. PolyphosphaZenes are Synthetic polymers, and aqueous 45 is generally the preferred temperature for Seeding and Solutions of polyphosphaZenes will gel in the presence of culture, especially with human cells. However, a wide range specific ions (Cohen et al., 1990). These polymers can be of temperatures are contemplated to be useful, ranging from used in the same manner as alginate. The exceedingly stable about 4 C. to approximately 50° C. The temperatures may backbone of these Synthetic polymerS allows significant be further optimized for different animals in veterinary alterations in Side-group functionality without losing the 50 procedures. gentle, physiologic gelling conditions (Allcock et al., 1988). The conditions of Seeding and matrix-cell maintenance 5. Synthetic Matrices that Mimic Natural Materials will preferably be optimized to enhance cell-matrix contact, There are advantages and disadvantages of both natural to maximize cell-matrix adhesion and to favor native tissue materials, e.g., collagens, and Synthetic materials, e.g., regeneration. Although certain variations in cell numbers, polyglycolic acids. Synthetic materials that incorporate 55 Seeding techniques, culture media and buffers, temperatures, design concepts or Specific biological activities of natural incubation times, and the like, may be advisable in certain biomaterials may combine the advantages of both types of circumstances, all Such variations will be routine to those of materials. The reproducible, large-scale Synthesis and flex skill in the art. It is well understood that certain parameters ible properties of Synthetic polymers can be combined with can be varied in order to improve the yield or activity of the biocompatibility and biological activity of natural mate 60 components in biological processes. Any Such variations rials. Such materials may be used in this invention. practiced in connection with regenerating oral tissueS eX Amino acid Sequences in ECM molecules that are respon Vivo using three dimensional matrices will thus be under Sible for Specific biological activities, e.g., cell binding, have stood to be encompassed within the present invention. been identified in recent years (Hynes, 1987; Hynes, 1990). As described above, different oral tissues will likely be This means researchers are able to design Synthetic materials 65 engineered using different starting cells that are appropriate that are capable of precise cellular interactions. Genetic for the tissue of interest, e.g., pulp-derived cells for pulp, engineering approaches are being utilized to prepare artifi gingival derived cells for gingival Submucosa. However, it 5,885,829 37 38 is also contemplated that cells not taken from the tissue of mass transport conditions may be required to ensure optimal interest will be useful, which cells are induced to express the development of oral tissues with appropriate Structure and appropriate phenotype, e.g., by exposure to Specific condi function following application of the regenerated tissue to tions in culture, via the introduction of foreign genes or an the oral tissue site. increased number of copies of natural Structural or regula AS the regenerated oral tissueS of the present invention tory genes, or combinations of Such methods. will be placed into an appropriate oral tissue A variety of criteria are available to assess the character microenvironment, the ability of the cells of the tissues to istics of the regenerated tissue. Using one or more of a range maintain a desired program of gene expression is not con of Structural and functional characteristics, the properties of sidered to pose a problem. However, if desired, any one of the regenerated tissues may be correlated with those of the a variety of delivery matrices that incorporate specific ECM natural oral tissues. By way of example only, one may molecules may be used to Supplement the correct Signaling mention: the gross histology and microstructure (using TEM to transplanted and/or the hosts own cells (Green et al., and SEM); the matrix composition and organization; the 1979; Yannas et al., 1981; Bell et al., 1981; Stern et al., 1990; cellular expression of Specific genes, at the level of gene Compton et al., 1989; Dixit, 1994; Kasai et al., 1994; transcription, mRNA levels, or the presence of Specific 15 Cavallaro et al., 1994, Anselme, 1992, Koide et al., 1993; proteins, e.g., membrane receptorS or matrix molecules, the Guenard et al., 1992). total cellularity and types of cells present, and the mechani Synthetic materials that incorporate Specific peptides to cal Strength and/or Stability of tissue. Techniques for carry enhance cell adhesion (McGrath et al., 1992; Barrera et al., ing out Such assays are routine and well known to those of 1993; Hubbell, 1993) may be used, including those that skill in the art. incorporate a variety of different peptides in order to mimic An exemplary, but by no means limiting, list of markers the multi-functional nature of ECM molecules (Hynes, which are contemplated for use in the present invention 1990). Growth factors promoting tissue development may includes: human gene markers for fibrous connective tissue be lacking or deficient in the host tissue site that the (produced by either pulp or gingival fibroblasts) for cellular engineered tissue is applied to. To address this concern, fibronectin or collagen type I, and III, measurable as protein 25 traditional controlled drug delivery technology may be inte or RNA, human gene markers for fibrous connective tissue grated with tissue engineering to provide transplanted cells (produced by either pulp or gingival fibroblasts) for BMP with Specific growth factors in their local environment. receptors BMPR-A1,-AII,-II or Act (activin) RI, BMP-2,-4 Mechanical signals are known to regulate the develop or -7, or MSX-2, a homeobox containing transcription factor ment of a variety of tissues, including muscle (Vandenburgh which has been implicated as a mediator of BMP signals et al., 1991), and bone (Carter et al., 1989). For example, during tissue (including tooth) development, measurable as engineered that are not Subjected to mechanical RNA and perhaps necessary to be responsible to BMP loading do not develop mechanical moduli as high as normal induction of bone or dentin formation; and alkaline phos tendons, even though they appear to be histologically iden phatase enzyme activity, which is associated with cells Such tical (Cao et al., 1994). Mechanical stimuli (e.g., Strain, as Osteoblasts and odontoblasts involved in mineralized 35 Shear) also clearly regulate the gene expression of cultured tissue formation. cells (Frangos, 1993). To engineer an optimally functional Additional markers include dentinsialposphophoryn oral tissue it may be necessary to provide the correct (DSPP), a putative specific marker for dentin, and bone mechanical Stimuli during the process of tissue develop Sialoprotein, osteonectin and Osteocalcin, which are highly ment. enriched in bone. Markers for use for the buccal mucosa 40 The mass transport between the engineered oral tissue and include cytokeratins 4 and 13. Further, markers for use in the host should be also considered. It will be understood that identification of taste buds include cytokeratins 8, 18 and 19, the metabolic requirements, e.g., Oxygen, of the developing which are present in taste buds but absent in the Surrounding tissue must be met, or the cells within the tissue will die. COS. 1. Growth Factors D. EXOgenous Factors 45 The use of growth factors in the context of cell prolifera If desired, protein growth factors that affect the prolifera tion and culture is generally well known in the art, although tion of cells and tissues may be used in conjunction with the growth factors, other than those naturally present in the engineering processes of the present invention. Likewise, Serum at low levels, have not been used in conjunction with cells that naturally elaborate Such factors and/or recombi oral tissue regeneration on a structural matrix eX Vivo. nant cells engineered to produce and Secrete Such factors 50 However, in that growth factors are routinely used in other may also be used to further stimulate the proliferation of contexts, one of skill in the art will readily understand how cells and tissues, or to direct the development of a given to apply growth factors in the context of the present inven tissue over another tissue when using Starter cells that have tion following a reading of the instant disclosure. the natural capacity to regenerate a number of different oral In general terms, it will be understood that a growth factor tissues. 55 that has already been established to have a beneficial physi It is preferable that the tissue-specific function of the cells ological effect on a particular cell type should be chosen for in the engineered oral tissue be maintained. The function of use in regenerating tissue containing Such cells. Certain cultured cells is Strongly dependent on the presence of growth factors may be used to Stimulate the proliferation of specific growth factors and ECM molecules (Stoker et al., a wide number of cell types, whereas other growth factors 1990). For example, cells can be switched from a phase of 60 may have a more limited or defined cell-specificity. Growth tissue-specific gene expression to one of proliferation simply factors may also be used to reduce Serum requirements, by altering the ECM presentation to the cell (Mooney et al., particularly where the use of Xenogenic Serum represents a 1992). problem. The microenvironment of an engineered tissue following The choice of a particular growth factor and cell combi transplantation may also be regulated during the process of 65 nation will be a routine matter for one of skill in the art. By tissue development, and perhaps beyond this time. Specific way of example only, one may mention platelet-derived ECM molecules, growth factors, mechanical Signals and/or growth factor, PDGF (such as PDGF-BB), which may be 5,885,829 39 40 used either alone or in combination with dexamethasone; front of the gene to be expressed, along with any necessary insulin-like growth factor I, basic fibroblast growth factor ribosome binding sites, RNA splice Sites, polyadenylation (bFGF); and epidermal growth factor (EGF). Site, and transcriptional terminator Sequences. For use in PDGF-BB and dexamethasone are effective for the mammalian cells, the control functions on the expression growth of pulp, periodontal ligament and gingival fibro vectors are often obtained from viral material. For example, blasts (Rutherford et al., 1992a, 1992b; 1993a), and are commonly used promoters are derived from polyoma, Aden particularly proposed for use in connection with these ovirus 2, and frequently, from Simian Virus 40 (SV40). The aspects of the invention. U.S. Pat. No. 5,149,691, incorpo early and late promoters of SV40 virus are particularly rated herein by reference, describes the use of combinations useful because both are obtained easily from the virus as a of PDGF and dexamethasone for the repair and regeneration fragment which also contains the SV40 viral origin of of tissues in vivo. U.S. Pat. Nos. 5,376,636 and 5,149,691, replication. It is also possible to utilize promoter or control each incorporated herein by reference, also describe the use Sequences normally associated with the desired gene of PDGF and glucocorticoids in tissue regeneration. Any Sequence, provided Such control Sequences are compatible Such teachings may be used in connection with the present with the host cell systems. invention. It is also known that this combination and PDGF/ 15 In mammalian cell vector Systems, the origin of replica IGF-1 induce regeneration of the periodontium in an animal tion may be obtained from either construction of the vector model of periodontitis (Rutherford et al., 1992b; 1993a). to include an exogenous origin, Such as may be derived from In addition to bFGF, another angiogenic factor that may SV40 or other viral (e.g., Polyoma, Adeno, VSV, BPV) be used is VEGF. Differentiation inducing factors are likely Source, or may be obtained from the host cell chromosomal to be useful, e.g., EGF and the like. Other hormones, such replication mechanism. If the vector is integrated into the as insulin, Steroids, and particularly, anti-inflammatory host cell chromosome, the latter is often Sufficient. agents are also contemplated for use here with. The use of exogenous growth factors to positively influ BMP proteins may be employed in certain aspects of the ence the engraftment, proliferation and Vascularization of present invention, such as those described in U.S. Pat. Nos. implanted cells and tissues is exemplified herein by the use 4,795,804; 4,877,864; 4.968,590; 5,011,691; 5,013,649; 25 of EGF, matrices and hepatocytes in liver regeneration 5,106,748; 5,108,753; 5,116,738; 5,141,905; 5,166,058 and Studies. 5,187,076, each incorporated herein by reference. For 3. Factors that Prevent or Inhibit Apoptosis example, the inventors have demonstrated that a single A number of factors are contemplated for use in the application of BMP-7 to a freshly and partially amputated present invention, based on their ability to block, prevent, or dental pulp induced reparative dentinogenesis in ferrets, reduce apoptosis. The factors contemplated for use with the monkeys and humans (Rutherford et al., 1993b, 1994, present invention include, but are not limited to, the follow 1995). Additionally, the inventors have demonstrated that ing compounds. The calcium ionophore A23.187 has been BMP-7 induced bone when implanted in gingiva, indicating shown to block apoptosis in certain Systems, Such as when that gingiva possess cells that are capable of forming min interleukin-3 (IL-3) is withdrawn from IL-3 dependent cells. eralized tissue Such as bone. 35 The thiol compounds pyrrolidine and dithiocarbamate have The growth factors or Stimulatory agents that are useful in also been shown to inhibit apoptosis. N-Acetyl-L-cysteine the context of the present invention may be purified from has been shown to prevent apoptotic death of neuronal cells natural Sources or may be recombinantly prepared proteins. (Ferrariet al., 1995) and TNF-C. induced apoptosis in U937 They may be obtained from commercial Sources, if desired. cells (Cossarizza et al., 1995). Nakajima et al. (1994) Those of skill in the art will know how to obtain and use 40 showed that actinomycin D, while a potent inducer of Such growth factors in the context of tissue regeneration in apoptosis in many cell lines, has been shown to Suppress light of the present disclosure. programmed cell death of PC12 cells induced by etoposide, 2. Cells that Produce Growth Factors an inhibitor of topoisomerase II These studies also showed It will be understood that the exogenous factors for use that cycloheximide, nerve growth factor and epidermal herewith may be produced by a population of cells that is 45 growth factor also rescued PC12 cells from etoposide co-cultured with the matrix-tissue and/or that is adminis induced death. Insulin-like growth factor-I (IGF-1) and the tered to the oral tissue of an animal with the tissue Sample IGF-1 receptor were also shown to inhibit etoposide-induced or matrix-tissue Sample The use of natural cells that elabo apoptosis in BALB/c 3T3 cells (Sell et al., 1995). rate any of the aforementioned or other growth factors, 3-Aminobenzamide has been shown to be an inhibitor of hormones or cytokines is thus contemplated. Such cells may 50 UV-induced apoptosis (Malorni et al., 1995). Aphidocolin be autologous cells that have also been proliferating in potentiates apoptosis induced by arabinosyl nucleosides in culture ex vivo. Certain cells, Such as T cells or B cells, may leukemia cell lines, and inhibits Vincristine-induced apop be enriched in the cell population, if desired. tosis in the p53-negative human prostate cancer cell line Equally, recombinant cells engineered to produce the PC-3 (Borner et al., 1995). L-AScorbic acid (vitamin C), growth factors, hormones, cytokines, hormone, neurotrans 55 catalase, follicle Stimulating hormone, N-acetyl-L-cysteine, mitters and the like may also be employed. Recombinant vasoactive intestinal peptide, cyclic GMP, hCG, interleukin engineering for protein production and Secretion, generally 1f8 (IL-1 B) and Superoxide dismutase have all been shown to using recombinant vectors, is now routine in the art, and is inhibit or SuppreSS apoptosis in cultured rat ovarian follicles described in further detail herein below. The culture of many (Flaws et al., 1995; Tilly and Tilly 1995; Chun et al., 1995). mammalian cells is, in itself, a routine procedure (Tissue 60 Aurintricarboxylic acid has been shown to inhibit apoptotic Culture, 1973, incorporated herein by reference). Examples cell death in various cell types induced by a variety of of useful host cell lines that may So engineered include, for factors (Benchokroun et al., 1995). example, VERO cells, HeLa cells, Chinese hamster ovary BAPTA/AM 1,2-bis(o-Aminophenoxy)ethane-N,N,N', (CHO) cell lines, and W138, BHK, COS-7, 293 and MDCK N-tetraacetic acid tetra (acetoxymethyl) ester inhibits cell lines. 65 thapSigargin-induced apoptosis in rat thymocytes (Jiang et Expression vectors for Such cells ordinarily include (if al., 1994). Caffeine has been shown to prevent apoptosis and necessary) an origin of replication, a promoter located in cell cycle effects induced by camptothecin and topotecan in 5,885,829 41 42 HL-60 cells (Traganos et al., 1993). Calpain inhibitor I factor (VEGF) or vascular permeability factor (VPF); mem inhibits apoptosis in thymocytes and metamyelocytes bers of the fibroblast growth factor family, including acidic (Squier et al., 1994), while leupeptin, calpain inhibitor II and fibroblast growth factor (aFGF) and basic fibroblast growth the E64 class of serine protease inhibitors have also been factor (bFGF); interleukin-8 (IL-8); epidermal growth factor shown to inhibit activation-induced programmed cell death (EGF); platelet-derived growth factor (PDGF) or platelet (Sarin et al., 1994). Cyclosporin A has been shown to derived endothelial cell growth factor (PD-ECGF); trans prevent anti-IgM and ionomycin-induced apoptosis in BLB forming growth factors alpha and beta (TGF-C, TGF-f); cell lines. tumor necrosis factor alpha (TNF-C.); hepatocyte growth The general Serine protease inhibitor 3,4- factor (HGF); granulocyte-macrophage colony Stimulating dichloroisocoumarin and the Specific thiol reagent N-ethyl factor (GM-CSF); insulin growth factor-1 (IGF-1); maleimide were shown to block apoptotic internucleosomal angiogenin, angiotropin, fibrin and nicotinamide (Folkman, DNA cleavage in thymocytes without the involvement of 1986, 1995; Auerbach and Auerbach, 1994; Fidler and Ellis, endonucleases (Cain et al., 1994). The cysteine protease 1994; Folkman and Klagsbrun, 1987; Nagy et al., 1995) inhibitors E64 and leupeptin, the calpain selective inhibitor 5. Cytokines acetyl-leucyl-leucyl-normethional, and the Serine protease 15 AS discussed above, in certain embodiments the use of inhibitors diisopropylfluorophosphate and phenylmethylsul particular cytokines in conjunction with the growth of oral fonyl fluoride were all shown to selectively block T-cell tissues is preferred. Table 4 below is an exemplary, but not receptor-triggered programmed cell death in murine T-cell limiting, list of additional cytokines and related factors hybridoma and in activated peripheral T-cells (Sarin et al., contemplated for use in the present invention. 1993). Tetrodotoxin, nimodipine, verapamil, flunarizine and R56865 all protect bovine chromaffin cells from veratridine TABLE 4 induced cell death (Maroto et al., 1994). Caspase inhibitors are also contemplated for use as apoptosis inhibitors. Cytokine Reference Forskolin and insulin growth factor-1 (IGF-1) both have human IL-1C. March et. al., Nature, 315:641, been shown to inhibit apoptosis in cerebellar granule cells, 25 985 although by distinct mechanisms (Galli et al., 1995). The murine IL-1C. Lomedico et al., Nature, 312:458, protein tyrosine kinase inhibitors genistein and herbimycin 984 human IL-1B March et al., Nature, 315:641, A have both been shown to prevent anti-CD3 monoclonal 985; Auron et al., Proc. Natl. antibody-induced thymic apoptosis (Migita et al., 1994). Acad. Sci. USA, 81:7907, 1984 Interleukin-6 (IL-6) inhibits constitutive, protein synthesis murine IL-1B Gray, J. Immunol., 137:3644, independent apoptosis of murine B-cell hybridoma 7TD1 986; Telford, NAR, 14:9955, 986 (Liu et al., 1994). The protein phosphatase inhibitors caly human IL-1 ra Eisenberg et al., Nature, 343:341, culin A and okadaic acid inhibit glucocorticoid-induced 990 apoptosis in T-cell hybridomas (Gjertsen et al., 1994), and human IL-2 Taniguchi et al., Nature, 302:305, calyculin A is known to prevent Y-radiation induced apop 35 983; Maeda et al., Biochem. Biophys. Res. Commun., 115: tosis in Burkitt's lymphoma cell line BM13674. 040, 1983 The protein kinase C activator phorbol-12-myristate-13 human IL-2 Taniguchi et al., Nature, 302:305, acetate inhibits apoptosis induced by the Fasantigen (Tepper 983 et al., 1995). 1-Pyrrolidinecarbodithioic acid prevents apo human IL-3 Yang et al., Cell, 47:3, 1986 murine IL-3 Yokota et al., Proc. Natl. Acad. ptosis in human promyeolocytic leukemia HL-60 cells and 40 Sci. USA, 81:1070, 1984: Fung et in thymocytes (Bessho et al., 1994). The calcium-channel al., Nature, 307:233, 1984; blockers nifedipine and niSoldipine, as well as the endonu Miyatake et al., Proc. Natl. Acad. clease inhibitor aurintricarboxylic acid have been shown to Sci. USA, 82:316, 1985 human IL-4 Yokota et al., Proc. Natl. Acad. block apoptosis in cultured human endothelial cells Sci. USA, 83:5894, 1986 (Escargueil-Blanc et al., 1997). Spermine has been shown to 45 murine IL-4 Norma et al., Nature, 319:640, inhibit morphological apoptosis, and the antioxidant thiore 1986; Lee et al., Proc. Natl. Acad. doxin inhibits apoptosis in Jurkat T-cells and human PBL Sci. USA, 83:2061, 1986 human IL-5 AZuma et al., Nuc. Acids Res., blasts (Sata et al., 1995). Additionally, the protease inhibi 14:9149, 1986 tors N'-Tosyl-L-Phe chloromethyl ketone, N'-Tosyl-L-Lys murine IL-5 Kinashi et al., Nature, 324:70, chloromethyl ketone, and to a lesser extent N'-Tosyl-L-Arg 50 1986; Mizuta et al., Growth methyl ester inhibit apoptosis in thymocytes (Bruno et al., Factors, 1:51, 1988 human IL-6 Hirano et al., Nature, 324:73, 1986 1992). murine IL-6 Van Snick et al., Eur. J. Immunol., 4. Factors that Promote Angiogenesis 18:193, 1988 The Successful engineering of new oral tissue requires the human IL-7 Goodwin et al., Proc. Natl. Acad. establishment of a vascular network in the new tissue. Case 55 Sci. USA, 86:302, 1989 murine IL-7 Namen et al., Nature, 333:571, reports of Successful tooth transplantation Suggest that pulp 1988 tissue may be capable of reestablishing vascular connections human IL-8 Schmid et al., J. Immunol. 139: (Myers and Fountain, 1974). However the inventors con 250, 1987: Matsushima et al., J. template the use of factors that promote angiogenesis to Exp. Med., 167:1883, 1988: Lindley et al., Proc. Natl. Acad. Support the neovascularization of the engineered oral tis 60 Sci. USA, 85:9199, 1988 SCS. human IL-9 Renauld et al., J. Immunol., 144: The induction of angiogenesis is mediated by a variety of 4235, 1990 factors, any of which may be used in conjunction with the murine IL-9 Renauld et al., J. Immunol., 144: 4235, 1990 present invention (Folkman and Klagsbrun, 1987, and ref human Angiogenin Kurachi et al., Biochemistry, 24: erences cited therein, each incorporated herein in their 65 5494, 1985 entirety by reference). Examples of angiogenic factors human GROC. Richmond et al., EMBO J., 7: includes, but is not limited to: Vascular endothelial growth 5,885,829 43 44

TABLE 4-continued TABLE 4-continued Cytokine Reference Cytokine Reference 2025, 1988 human Calcitonin Le Moullec et al., FEBS Lett., 167: murine MIP-1C. Davatelis et al., J. Exp. Med., 167: 93, 1984 1939, 1988 human Hirudin (E. coli optimized) Dodt et al., FEBS Lett., 165:180, murine MIP-1B Sherry et al., J. Exp. Med., 168: 1984 2251, 1988 human MIF Weiser et al., Proc. Natl. Acad. Sci. USA, 86:7522, 1989 1O E. Clinical Applications human G-CSF Nagata et al., Nature, 319:415, Oral tissues engineered in accordance with the present 1986: Souza et al., Science, 232: 61, 1986 invention have evident clinical utility in the regeneration of human GM-CSF Cantrell et al., Proc. Natl. Acad. oral tissues and structures in Vivo. Inducing or Speeding up Sci. USA, 82:6250, 1985; Lee et the overall regeneration process by administering regener al., Proc. Natl. Acad. Sci. USA, 15 ated tissue to the Site of injury is beneficial as it lessens the 82:4360, 1985; Wong et al., Science, 228:810, 1985 chance that the body will attempt to repair the injury, leading murine GM-CSF Gough et al., EMBO J., 4:645, to the formation of Scar tissue and the loSS of normal tissue 1985 Structure and function. human M-CSF Wong, Science, 2.35:1504, 1987; Tissues engineered using autologous cells will likely be Kawasaki, Science, 230; 291, 1985; Ladner, EMBO J., 6:2693, those first used in a clinical Setting. In a Specific embodiment 1987 of the invention, dental pulp tissues have already been human EGF Smith et al., Nuc. Acids Res., 10: regenerated. Such dental pulp tissues will be useful in 4467, 1982: Bell et al., NAR, 14: effecting the repair of dental pulp and reparative dentin 8427, 1986 human TGF-C. Derynck et al., Cell, 38:287, 1984 formation following disease or Surgical manipulation. human FGF acidic Jaye et al., Science, 233:541, 1986; 25 The regenerated dental pulp tissues were made by isolat Gimenez-Gallego et al., Biochem. ing fibroblasts from human dental pulp and multiplying Biophys. Res. Commun., 138:611, them in culture. These cells were Seeded onto Synthetic 986; Harper et al., Biochem., 25:4097, 1986 extracellular matrices fabricated from fibers (approximately human B-ECGF Jaye et al., Science, 233:541, 1986 15um in diameter) of polyglycolic acid (PGA). The pulp human FGF basic Abraham et al., EMBO J., 5:2523, derived cells adhered to the fibers and proliferated over time. 986: Sommer et al., Biochem. Biophys. Res. Comm., 144:543, New tissues were formed over 60 days in culture that 987 histologically resembled native dental pulp tissue. These murine IFN-B Higashi et al., J. Biol. Chem., new tissues also had a cellularity in the same range as adult 258:9522, 1983; Kuga, NAR, human pulp. Immunohistochemical and histochemical 7:3291, 1989 35 analysis of the extracellular matrix revealed that it contained human IFN-y Gray et al., Nature, 295:503, 982: Devos et al., NAR, 10:2487, type I collagen and cellular fibronectin; normal constituents 982: Rinderknecht, J. Biol. of mature dental pulp tissue. Chem., 259:6790, 1984 Although the present tissue regeneration and transplanta human IGF-I Jansen et al., Nature, 306:609, tion methods are considered to be effective in and of 983; Rotwein et al., J. Biol. Chem., 261:4828, 1986 40 themselves, the application of the regenerated tissue may be human IGF-II Bell et al., Nature, 310:775, 1984 combined with growth factors (e.g., Folkman and human B-NFG chain Ullrich et al., Nature, 303:821, Klagsbrun, 1987) in certain applications, e.g., to stimulate blood vessel formation (angiogenesis) or to promote cell human PDGFA chain Betsholtz et al., Nature, 320:695, 986 growth. Angiogenesis can also be promoted by controlling human PDGFB chain Johnsson et al., EMBO J., 3:921, 45 the microstructure of the matrix (Mooney et al., 1994b; 984; Collins et al., Nature, 316: Mikos et al., 1993b). 748, 1985 Direct local application of recombinant growth factors human TGF-B1 Derynck et al., Nature, 316:701, 985 (e.g., BMP-2) has been shown to induce reparative denti human TNF-C. Pennica et al., Nature, 312:724, nogenesis in dogs and primates when placed on partially 984; Fransen et al., Nuc. Acids 50 amputated dental pulps (Rutherford et. al., 1993b; Res., 13:4417, 1985 human TNF-B Gray et al., Nature, 312:721, 1984 Nakashima, 1994; Rutherford et. al., 1994), or on a freshly murine TNF-B Gray et al., Nucl. Acids Res., 15: cut dental Surface ("transdentinal” application, Rutherford 3937, 1987 et. al., 1995). However, in many clinical situations no pulp human E-Selectin Bevilacqua et al., Science, 243: remains to Stimulate. The present invention provides new, 1160, 1989; Hensley et al., J. Biol. 55 preferably autologous, pulp tissue to replace lost pulp tissue Chem., 269:23949, 1994 human ICAM-1 Simmons et al., Nature, 331:624, and form reparative dentin. 1988 AS an example, autologous pulp-derived fibroblasts are human PECAM Simmons et al., J. Exp. Med., 171: obtained from healthy molars or wisdom teeth and are 2147, 1990 expanded in culture to engineer the new pulp tissues for use human VCAM-1 Hession et al., J. Biol. Chem., 266: 6682: Osborn et al., Cell, 59:1203, 60 in diseased teeth. Alternatively, tissue extracted from the 1989 original diseased tooth may itself be used to regenerate human L-Selectin (membrane bound) Ord et al., J. Biol. Chem..., 265: dental pulp by expanding and culturing the viable cells that 7760, 1990; Tedder et al., J. Exp. remained in the extracted material. Med., 170:123, 1989 human L-Selectin (soluble form) Ord et al., J. Biol. Chem..., 265: Ultimately, the Successful engineering of new oral tissue 7760, 1990; Tedder et al., J. Exp. 65 requires the establishment of a vascular network in the new Med., 170:123, 1989 tissue and the development of an appropriate Synthetic Scaffold for cell transplantation. Case reports of Successful 5,885,829 45 46 tooth transplantation Suggest that pulp tissue may be capable chon (1992); Barile (1994); Black (1992); Kumar et al., of reestablishing vascular connections (Myers and Fountain, (1992); and Darnell et al., (1990): each incorporated herein 1974) and reinnervating (Holland and Robinson, 1987). by reference. The demonstration that neovascularization and reinner The ADA Specifications are guidelines that govern the Vation can occur in transplanted teeth with intact pulps materials and devices used in dentistry (restorative dental argues that these components of tissue repair may readily materials are defined by the FDA as “devices”). The phi occur in the pulp tissue developed ex vivo. Such tissues will losophy requires that a tiered System be used, Starting with be leSS dense and therefore more easily penetrated than fully initial tests (in vitro and in vivo), followed by secondary mature dental pulp. Additionally whereas revascularization tests (more involved in Vivo tests), and then usage tests is essential for Survival of the implanted tissue the same case which place the material into actual use, usually in higher has not been and is likely not true for reinnervation. animals (monkeys or dogs). Therefore, the regenerated pulp tissue of the present inven Many of the tests in the ADA specification are out of date, tion represent an ideal Starting material for the Successful however, they are still being used. The ADA Specification engineering of new oral tissue. currently relies heavily on animal testing. The present inven The importance and application of other regenerated oral 15 tion provides valuable alternatives to the out-dated methods tissueS of the present invention will be apparent to those of of the initial tests and should also reduce the amount of skill in the art in light of the present disclosure. For example, animal testing to a minimum, as the initial tests are more the present invention's use in engineering periodontal tissue relevant when performed a three dimensional native-like is important as periodontal disease is one of the most oral tissue. Significant oral health problems in the USA. Approximately Using the invention's system for in vitro biocompatibility 35% of the U.S. population is estimated to have periodontitis testing, one of skill in the art would be able to define an and 80% of these also suffer from gingivitis. unacceptable level of toxicity in order to identify an agent as The generation of new periodontal tissue occurs by using unsafe for human and/or veterinary use. Acceptable levels of Staring cells from the periodontal ligament or gingival no or minimal toxicity would identify an agent as Safe for Submucosa, as described above. After tissue growth eX Vivo, 25 human and/or veterinary use. If necessary, results from the the regenerated tissue would be applied to the exposed root present in vitro biocompatibility test could be initially Surface and immediately adjacent tissues using the Modified correlated with the ADA Specifications and Stan Widman approach. It is contemplated that a combination of dards to yield results more instantly interpretable by those of tissue and growth factor delivery will provide a particularly skill in the art. advantageous means of regenerating a healthy periodon G. Engineering of Cells tium. AS described herein above, recombinant cells engineered It will also be understood that for most oral applications to produce a variety of exogenous factors, hormones, the use of antimicrobials/antibiotics may be important in cytokines, hormone, neurotransmitters and the like are con order to minimize infection of the tissue during development templated for use in the present invention. Expression vec and to resolve certain oral diseases. The local or Systemic 35 tors for Such cells ordinarily include (if necessary) an origin administration of antimicrobials in connection with the of replication, a promoter located in front of the gene to be present invention is thus contemplated. expressed, along with any necessary ribosome binding sites, Antibiotics of choice for common pathogens include, e.g., RNA splice Sites, polyadenylation site, and transcriptional penicillins, amplicillin, amoxicillin, erythromycin, terminator Sequences. Once the desired vector construct is cephalosporins, tetracyclines and the like. AntibioticS Such 40 obtained, it may be delivered into the desired cells by a as these may be used clinically at a range of doses, as is number of different techniques. known to those of Skill in the art. Depending on the 1. Promoters and Enhancers circumstances, antimicrobial agents will preferably be used Recombinant vectors form important further aspects of orally, although parenteral treatment regimens are not the present invention. The term "expression vector or con excluded. Appropriate doses are well known to those of Skill 45 Struct” means any type of genetic construct containing a in the art, and are further described in various publications, nucleic acid coding for a gene product in which part or all such as Reese and Betts (1993), incorporated herein by of the nucleic acid encoding Sequence is capable of being reference. Table 5 and Table 6 of Reese and Betts (1993) are transcribed. The transcript may be translated into a protein, particularly incorporated herein by reference to provide but it need not be. Thus, in certain embodiments, expression ready reference to the currently recommended doses of a 50 includes both transcription of a gene and translation of a variety of antimicrobial agents. RNA into a gene product. In other embodiments, expression F. Screening ASSayS only includes transcription of the nucleic acid, for example, The regenerated oral tissueS obtained from the present to generate antisense constructs. invention may also be used as novel Systems for testing the Particularly useful vectors are contemplated to be those toxicity and/or biocompatibility of materials and chemicals 55 vectors in which the coding portion of the DNA segment, intended for use in dentistry and oral medicine. whether encoding a full length protein or Smaller peptide, is In Such novel Systems, the three-dimensional human positioned under the transcriptional control of a promoter. A tissueS obtained from the invention represent a more physi “promoter” refers to a DNA sequence recognized by the ologically relevant System than Standard two-dimensional Synthetic machinery of the cell, or introduced Synthetic cell culture models currently utilized for in vitro testing of 60 machinery, required to initiate the Specific transcription of a biocompatibility (Schmaltz, 1994). gene. The phrases “operatively positioned”, “under control” In general, one may use a three-dimensional oral tissue of or “under transcriptional control’ means that the promoter is the invention to test for the cytotoxicity, mutagenicity and/or in the correct location and orientation in relation to the neoplastic transformation of cells exposed to a variety of nucleic acid to control RNA polymerase initiation and agents or combinations thereof. The following texts describe 65 expression of the gene. suitable methods that exemplify those that may be used in The promoter may be in the form of the promoter that is connection with this invention: von Recum (1986); Ecobi naturally associated with a particular gene, as may be 5,885,829 47 48 obtained by isolating the 5' non-coding Sequences located to act over a large distance had little precedent in classic upstream of the coding Segment or exon, for example, using Studies of prokaryotic transcriptional regulation. Subsequent recombinant cloning and/or PCR technology, in connection work showed that regions of DNA with enhancer activity are with the compositions disclosed herein (PCR technology is organized much like promoters. That is, they are composed disclosed in U.S. Pat. No. 4,683.202 and U.S. Pat. No. 5 of many individual elements, each of which binds to one or 4,682,195, each incorporated herein by reference). more transcriptional proteins. In other embodiments, it is contemplated that certain The basic distinction between enhancers and promoters is advantages will be gained by positioning the coding DNA operational. An enhancer region as a whole must be able to Segment under the control of a recombinant, or Stimulate transcription at a distance; this need not be true of heterologous, promoter. AS used herein, a recombinant or promoter region or its component elements. On the other heterologous promoter is intended to refer to a promoter that hand, a promoter must have one or more elements that direct is not normally associated with a particular gene in its initiation of RNA synthesis at a particular site and in a natural environment. Such promoters may include promot particular orientation, whereas enhancers lack these speci erS normally associated with other genes, and/or promoters ficities. Promoters and enhancers are often overlapping and isolated from any other bacterial, Viral, eukaryotic, or mam 15 contiguous, often Seeming to have a very Similar modular malian cell. organization. Naturally, it will be important to employ a promoter that Tables 5 and 6 below list several elements/promoters effectively directs the expression of the DNA segment in the which may be employed, in the context of the present Selected cell type chosen for expression. The use of pro invention, to regulate gene expression. This list is not moter and cell type combinations for protein expression is intended to be exhaustive of all the possible elements generally known to those of skill in the art of molecular involved in the promotion of transgene expression but, biology, for example, see Sambrook et al. (1989), incorpo merely, to be exemplary thereof. Additionally any promoter/ rated herein by reference. The promoters employed may be enhancer combination (as per the Eukaryotic Promoter Data constitutive, or inducible, and can be used under the appro Base EPDB) could also be used to drive expression of a priate conditions to direct high level expression of the 25 transgene. Use of a T3, T7 or SP6 cytoplasmic expression introduced DNA segment. System is another possible embodiment. Eukaryotic cells can At least one module in a promoter functions to position Support cytoplasmic transcription from certain bacterial pro the start site for RNA synthesis. The best known example of moters if the appropriate bacterial polymerase is provided, this is the TATA box, but in some promoters lacking a TATA either as part of the delivery complex or as an additional box, Such as the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the genetic expression construct. SV40 late genes, a discrete element overlying the Start Site itself helps to fix the place of initiation. TABLE 5 Additional promoter elements regulate the frequency of Inducible Elements transcriptional initiation. Typically, these are located in the 35 region 30-110 bp upstream of the Start Site, although a Element Inducer References number of promoters have been shown to contain functional MT II Phorbol Ester (TFA) Palmiter et al., 1982: elements downstream of the Start Site as well. The Spacing Heavy metals Haslinger and Karin, between promoter elements frequently is flexible, So that 1985; Searle et al., 1985; Stuart et al., 1985: promoter function is preserved when elements are inverted 40 Imagawa et al., 1987: or moved relative to one another. In the tk promoter, the Karin (R), 1987; Angel et spacing between promoter elements can be increased to 50 al., 1987b; McNeal et bp apart before activity begins to decline. Depending on the al., 1989 MMTV (mouse Glucocorticoids Huang et al., 1981; Lee promoter, it appears that individual elements can function mammary tumor virus) et al., 1981; Majors and either co-operatively or independently to activate transcrip 45 Varmus, 1983; Chandler tion. et al., 1983; Lee et al., The particular promoter that is employed to control the 1984; Fonta et al., 1985; Sakai et al., 1986 expression of a nucleic acid is not believed to be critical, So B-Interferon poly(rDX Tavernier et al., 1983 long as it is capable of expressing the nucleic acid in the poly(rc) targeted cell. Thus, where a human cell is targeted, it is 50 Adenovirus 5 E2 Ela Imperiale and Nevins, preferable to position the nucleic acid coding region adja 1984 Collagenase Phorbol Ester (TPA) Angle et al., 1987a cent to and under the control of a promoter that is capable Stromelysin Phorbol Ester (TPA) Angle et al., 1987b of being expressed in a human cell. Generally speaking, SV40 Phorbol Ester (TFA) Angel et al., 1987b Such a promoter might include either a human or viral Murine MX Gene Interferon, Newcastle promoter. 55 Disease Virus GRP78 Gene A231.87 Resendez et al., 1988 In various other embodiments, the human cytomegalovi C-2-Macroglobulin IL-6 Kunz et al., 1989 rus (CMV) immediate early gene promoter, the SV40 early Vimentin Serum Rittling et al., 1989 promoter and the Rous Sarcoma virus long terminal repeat MHC Class I Gene H Interferon Blanar et al., 1989 can be used to obtain high-level expression of transgenes. 2kb HSP70 Ela, SV40 Large T Taylor et al., 1989; The use of other viral or mammalian cellular or bacterial 60 Antigen Taylor and Kingston, phage promoters which are well-known in the art to achieve 1990a,b expression of a transgene is contemplated as well, provided Proliferin Phorbol Ester-TPA Mordacq and Linzer, that the levels of expression are Sufficient for a given 1989 Tumor Necrosis Factor FMA Hensel et al., 1989 purpose. Thyroid Stimulating Thyroid Hormone Chatterjee et al., 1989 Enhancers were originally detected as genetic elements 65 Hormone C. Gene that increased transcription from a promoter located at a distant position on the same molecule of DNA. This ability 5,885,829 49 SO

TABLE 6 TABLE 6-continued Other Promoter/Enhancer Elements Other Promoter/Enhancer Elements Promoter/Enhancer References Promoter/Enhancer References Immunoglobulin Heavy Chain Hanerji et al., 1983; Gilles et al., Spalholz et al., 1985; Lusky and 1983: Grosschedl and Baltimore, Botchan, 1986; Cripe et al., 1987: 1985; Atchinson and Perry, 1986, Gloss et al., 1987: Hirochika et al., 1987: Imler et al., 1987; Weinberger 1987, Stephens and Hentschel, 1987: et al., 1988; Kiledjian et al., 1988; 1O Glue et al., 1988 Porton et al., 1990 Hepatitis B Virus Bulla and Siddiqui, 1986; Jameel and Immunoglobulin Light Chain Queen and Baltimore, 1983; Siddiqui, 1986; Shaul and Ben-Levy, Picard and Schaffner, 1984 1987; Spandau and Lee, 1988; T-Cell Receptor Luria et al., 1987, Winoto and Vannice and Levinson, 1988 Baltimore, 1989; Redondo et al., Human Immunodeficiency Virus Muesing et al., 1987; Hauber and 1990 15 Cullan, 1988; Jakobovits et al., 1988: HLA DQ C. and DQB Sullivan and Peterlin, 1987 Feng and Holland, 1988; Takebe et B-Interferon Goodbourn et al., 1986; Fujita et al., al., 1988: Rowen et al., 1988: 1987: Goodbourn and Manilatis, 1985 Berkhout et al., 1989; Laspia et al., Interleukin-2 Greene et al., 1989 1989; Sharp and Marciniak, 1989; Interleukin-2 Receptor Green et al., 1989; Lin et al., 1990 Braddock et al., 1989 MHC Class II 5 Koch et al., 1989 Cytomegalovirus Weber et al., 1984: Boshart et al., MHC Class II HLA-DRC. Sherman et al., 1989 1985; Foecking and Hofstetter, 1986 3-Actin Kawamoto et al., 1988; Ng et al., Gibbon Ape Leukemia Virus Holbrook et al., 1987: Quinn et al., 1989 1989 Muscle Creatine Kinase Jaynes et al., 1988; Horlick and Benfield, 1989; Johnson et al., 1989a Prealbumin (Transthyretin) Costa et al., 1988 2. Marker Genes Elastase I Omitz et al., 1987 25 The present invention also provides recombinant candi Metallothionein Karin et al., 1987: Culotta and Hamer, 1989 date Screening and Selection methods which are based upon Collagenase Pinkert et al., 1987; Angel et al., whole cell assays and which, preferably, employ a reporter 1987 gene that conferS on its recombinant hosts a readily detect Albumin Gene Pinkert et al., 1987, Tronche et al., able phenotype that emerges only under conditions where a 1989, 1990 general DNA promoter positioned upstream of the reporter C-Fetoprotein Godbout et al., 1988; Campere and Tilghman, 1989 gene is functional. Generally, reporter genes encode a t-Globin Bodine and Ley, 1987; Perez-Stable polypeptide (marker protein) not otherwise produced by the and Constantini, 1990 host cell which is detectable by analysis of the cell culture, B-Globin Trudel and Constantini, 1987 e.g., by fluorometric, radioisotopic or spectrophotometric e-fos Cohen et al., 1987 35 analysis of the cell culture. c-HA-ras Triesman, 1986; Deschamps et al., 1985 In other aspects of the present invention, a genetic marker Insulin Edlund et al., 1985 is provided which is detectable by Standard genetic analysis Neural Cell Adhesion Molecule Hirsch et al., 1990 techniques, such as DNA amplification by PCRTM or hybrid (NCAM) ization using fluorometric, radioisotopic or spectrophoto 1-Antitrypain Latimer et al., 1990 H2B (TH2B) Histone Hwang et al., 1990 40 metric probes. Mouse or Type I Collagen Ripe et al., 1989 a. Screening Glucose-Regulated Proteins Chang et al., 1989 Exemplary enzymes include esterases, phosphatases, pro (GRP94 and GRP78) teases (tissue plasminogen activator or urokinase) and other Rat Growth Hormone Larsen et al., 1986 Human Serum Amyloid A (SAA) Edbrooke et al., 1989 enzymes capable of being detected by their activity, as will Troponin I (TN I) Yutzey et al., 1989 45 be known to those skilled in the art. Contemplated for use in Platelet-Derived Growth Factor Pech et al., 1989 the present invention is green fluorescent protein (GFP) as a Duchenne Muscular Dystrophy Klamut et al., 1990 marker for transgene expression (Chalfie et al., 1994). The SV40 Banerji et al., 1981; Moreau et al., 1981; Sleigh and Lockett, 1985; Firak use of GFP does not need exogenously added Substrates, and Subramanian, 1986: Herr and only irradiation by near UV or blue light, and thus has Clarke, 1986: Imbra and Karin, 1986: 50 Significant potential for use in monitoring gene expression in Kadesch and Berg, 1986; Wang and living cells. Calame, 1986: Ondek et al., 1987: Kuhl et al., 1987 Schaffner et al., Other particular examples are the enzyme chlorampheni 988 col acetyltransferase (CAT) which may be employed with a Polyoma Swartzendruber and Lehman, 1975: radiolabelled Substrate, firefly and bacterial luciferase, and Vasseur et al., 1980: Katinka et al., 55 the bacterial enzymes B-galactosidase and B-glucuronidase. 980, 1981; Tyndell et al., 1981; Dandolo et al., 1983; deVilliers et Other marker genes within this class are well known to those al., 1984; Hen et al., 1986; Satake of skill in the art, and are Suitable for use in the present et al., 1988; Campbell and Villarreal, invention. 988 b. Selection Retroviruses Kriegler and Botchan, 1982, 1983; Levinson et al., 1982; Kriegler et al., 60 Another class of reporter genes which confer detectable 983, 1984a,b, 1988: Bosze et al., characteristics on a host cell are those which encode 986; Miksicek et al., 1986: Celander polypeptides, generally enzymes, which render their trans and Haseltine, 1987: Thiesen et al., formants resistant against toxins. Examples of this class of 988; Celander et al., 1988: Chol et al., 1988: Reisman and Rotter, 1989 reporter genes are the neogene (Colberre-Garapin et al., Papilloma Virus Campo et al., 1983; Lusky et al., 65 1981) which protects host cells against toxic levels of the 983; Spandidos and Wilkie, 1983; antibiotic G418, the gene conferring Streptomycin resistance (U.S. Pat. No. 4,430,434), the gene conferring hygromycin 5,885,829 S1 52 B resistance (Santerre et al., 1984; U.S. Pat. Nos. 4,727,028, Oligonucleotides which contain C-5 propyne analogues of 4,960,704 and 4.559,302), a gene encoding dihydrofolate uridine and cytidine have been shown to bind RNA with reductase, which confers resistance to methotrexate (Alt et high affinity and to be potent antisense inhibitors of gene al., 1978), the enzyme HPRT, along with many others well expression. known in the art (Kaufman, 1990). 4. Ribozymes 3. AntiSense Another method for inhibiting gene expression contem AntiSense methodology takes advantage of the fact that plated in the present invention is via ribozymes. Although nucleic acids tend to pair with “complementary' Sequences. proteins traditionally have been used for catalysis of nucleic By complementary, it is meant that polynucleotides are those acids, another class of macromolecules has emerged as which are capable of base-pairing according to the Standard useful in this endeavor. Ribozymes are RNA-protein com Watson-Crick complementarity rules. That is, the larger plexes that cleave nucleic acids in a Site-specific . purines will base pair with the Smaller pyrimidines to form Ribozymes have Specific catalytic domains that possess combinations of guanine paired with cytosine (G.C) and endonuclease activity (Kim and Cech, 1987; Gerlach et al., adenine paired with either thymine (A:T) in the case of 1987; Forster and Symons, 1987). For example, a large DNA, or adenine paired with uracil (AU) in the case of 15 number of ribozymes accelerate phosphoester transfer reac RNA. Inclusion of leSS common bases Such as inosine, tions with a high degree of Specificity, often cleaving only 5-methylcytosine, 6-methyladenine, hypoxanthine and oth one of Several phosphoesters in an oligonucleotide Substrate erS in hybridizing Sequences does not interfere with pairing. (Cech et al., 1981; Michel and Westhof, 1990; Reinhold Targeting double-stranded (ds) DNA with polynucle Hurek and Shub, 1992). This specificity has been attributed otides leads to triple-helix formation; targeting RNA will to the requirement that the Substrate bind via Specific base lead to double-helix formation. AntiSense polynucleotides, pairing interactions to the internal guide Sequence (“IGS) when introduced into a target cell, Specifically bind to their of the ribozyme prior to chemical reaction. target polynucleotide and interfere with transcription, RNA Ribozyme catalysis has primarily been observed as part of processing, transport, translation and/or Stability. AntiSense Sequence-specific cleavage/ligation reactions involving RNA constructs, or DNA encoding such antisense RNAS, 25 nucleic acids (Joyce, 1989; Cech et al., 1981). For example, may be employed to inhibit gene transcription or translation U.S. Pat. No. 5,354,855 reports that certain ribozymes can or both within the cells of the present invention. act as endonucleases with a Sequence Specificity greater than AntiSense constructs may be designed to bind to the that of known ribonucleases and approaching that of the promoter and other control regions, exons, introns or even DNA restriction enzymes. Thus, Sequence-specific exon-intron boundaries of a gene. It is contemplated that ribozyme-mediated inhibition of gene expression may be effective antisense constructs will often include regions particularly Suited to therapeutic applications (Scanlon et al., complementary to intron/exon Splice junctions. Thus, anti 1991; Sarver et al., 1990; Sioud et al., 1992). Recently, it Sense constructs with complementarity to regions within Was reported that ribozymes elicited genetic changes in 50–200 bases of an intron-exon Splice junction are contem Some cells lines to which they were applied; the altered plated for use herewith. It has been observed that some exon 35 genes included the oncogenes H-ras, c-foS and genes of HIV. Sequences can be included in the construct without Seriously Most of this work involved the modification of a target affecting the target Selectivity thereof. The amount of exonic mRNA, based on a specific mutant codon that is cleaved by material included will vary depending on the particular exon a specific ribozyme. and intron Sequences used. One can readily test whether too Several different ribozyme motifs have been described much exon DNA is included simply by testing the constructs 40 with RNA cleavage activity (Symons, 1992). Examples that in vitro to determine whether the expression of genes having are expected to function equivalently for the down regula complementary Sequences is affected. tion of gene expression include Sequences from the Group I "AntiSense' or “complementary means polynucleotide Self Splicing introns including Tobacco Ringspot Virus Sequences that are Substantially complementary over their (Prody et al., 1986), Avocado Sunblotch Viroid (Palukaitis et entire length and have very few base mismatches. For 45 al., 1979; Symons, 1981), and Lucerne Transient Streak example, Sequences of fifteen bases in length may be termed Virus (Forster and Symons, 1987). Sequences from these complementary when they have complementary nucleotides and related viruses are referred to as hammerhead ribozyme at thirteen or fourteen positions. Naturally, Sequences which based on a predicted folded Secondary Structure. are completely complementary will be sequences which are Other suitable ribozymes include sequences from RNase entirely complementary throughout their entire length and 50 P with RNA cleavage activity (Yuan et al., 1992, Yuan and have no base mismatches. Other Sequences with lower Altman, 1994, U.S. Pat. Nos. 5,168,053 and 5,624,824), degrees of homology also are contemplated. For example, an hairpin ribozyme structures (Berzal-Herranz et al., 1992; antisense construct which has limited regions of high Chowrira et al., 1993) and Hepatitis Delta virus based homology, but also contains a non-homologous region (e.g., ribozymes (U.S. Pat. No. 5,625,047). The general design and ribozyme) could be designed. These molecules, though 55 optimization of ribozyme directed RNA cleavage activity having less than 50% homology, would bind to target has been discussed in detail (Haseloff and Gerlach, 1988, Sequences under appropriate conditions. Symons, 1992, Chowrira et al., 1994; Thompson et al., It may be advantageous to combine portions of genomic 1995). DNA with cDNA or synthetic sequences to generate specific The other variable on ribozyme design is the selection of constructs. For example, where an intron is desired in the 60 a cleavage Site on a given target RNA. Ribozymes are ultimate construct, a genomic clone will need to be used. targeted to a given Sequence by Virtue of annealing to a site The cDNA or a synthesized polynucleotide may provide by complimentary base pair interactions. Two Stretches of more convenient restriction sites for the remaining portion homology are required for this targeting. These Stretches of of the construct and, therefore, would be used for the rest of homologous Sequences flank the catalytic ribozyme struc the Sequence. In certain embodiments, one may wish to 65 ture defined above. Each Stretch of homologous Sequence employ antisense constructs which include other elements, can vary in length from 7 to 15 nucleotides. The only for example, those which include C-5 propyne pyrimidines. requirement for defining the homologous Sequences is that, 5,885,829 S3 S4 on the target RNA, they are separated by a specific Sequence charge to generate an electrical current, which in turn which is the cleavage Site. For hammerhead ribozyme, the provides the motive force (Yang et al., 1990). Another cleavage Site is a dinucleotide Sequence on the target RNA method involves the use of a Biolistic Particle Delivery is a uracil (U) followed by either an adenine, cytosine or System, which can be used to propel particles coated with uracil (A.C or U) (Perriman et al., 1992; Thompson et al., DNA through a Screen, Such as StainleSS Steel or NyteX 1995). The frequency of this dinucleotide occurring in any Screen, onto a filter Surface covered with cells in Suspension. given RNA is statistically 3 out of 16. Therefore, for a given The Screen disperses the particles So that they are not target messenger RNA of 1000 bases, 187 dinucleotide delivered to the recipient cells in large aggregates. It is cleavage Sites are Statistically possible. believed that a Screen intervening between the projectile Designing and testing ribozymes for efficient cleavage of apparatus and the cells to be bombarded reduces the size of a target RNA is a process well known to those skilled in the projectile aggregates and may contribute to a higher fre art. Examples of Scientific methods for designing and testing quency of transformation by reducing the damage inflicted ribozymes are described by Chowrira et al., (1994) and on the recipient cells by projectiles that are too large. Lieber and Strauss (1995), each incorporated by reference. For the bombardment, cells in Suspension are preferably The identification of operative and preferred Sequences for 15 concentrated on filters, or alternatively on Solid culture use in targeted ribozymes is simply a matter of preparing and medium. The cells to be bombarded are positioned at an testing a given Sequence, and is a routinely practiced appropriate distance below the macroprojectile Stopping “screening” method known to those of skill in the art. plate. If desired, one or more Screens are also positioned 5. DNA Delivery between the acceleration device and the cells to be bom In order to effect expression of a gene construct, the barded. expression construct must be delivered into a cell. Several In bombardment transformation, one may optimize the methods for the transfer of genetic constructs into cells are prebombardment culturing conditions and the bombardment contemplated by the present invention. The particular parameters to yield the maximum numbers of Stable trans method employed may depend upon when the DNA con formants. Both the physical and biological parameters for Struct is to be transferred into the cells. It is generally 25 bombardment are important in this technology. Physical preferred to introduce the DNA segment to the cells before factors are those that involve manipulating the DNA/ Seeding the cells onto the matrix. In one embodiment of the microprojectile precipitate or those that affect the flight and present invention, the expression construct may consist only Velocity or either the macro- or microprojectiles. Biological of naked recombinant DNA or plasmids. Transfer of the factors include all Steps involved in manipulation of cells construct may be performed by any of the methods men before and immediately after bombardment, the osmotic tioned which physically or chemically permeabilize the cell adjustment of target cells to help alleviate the trauma asso membrane. ciated with bombardment, and also the nature of the trans a. Electroporation forming DNA, Such as linearized DNA or intact Supercoiled In certain preferred embodiments of the present invention, plasmids. It is believed that pre-bombardment manipulations the genetic construct is introduced into cells via electropo 35 are especially important for Successful transformation of ration. Electroporation involves the exposure of a Suspen primordial germ cells. Sion of cells and DNA to a high-voltage electric discharge. Accordingly, it is contemplated that one may wish to Transfection of eukaryotic cells using electroporation has adjust various of the bombardment parameters in Small Scale been quite Successful. Mouse pre-B lymphocytes have been Studies to fully optimize the conditions. One may particu transfected with human kappa-immunoglobulin genes 40 larly wish to adjust physical parameterS Such as gap (Potter et al., 1984), and rat hepatocytes have been trans distance, flight distance, tissue distance and helium pressure. fected with the chloramphenicol acetyltransferase gene One may also optimize the trauma reduction factors by (Tur-Kaspa et al., 1986) in this manner. modifying conditions which influence the physiological It is contemplated that electroporation conditions for cells State of the recipient cells and which may therefore influence from different Sources may be optimized. One may particu 45 transformation and integration efficiencies. For example, the larly with to optimize Such parameters as the Voltage, the oSmotic State, tissue hydration and the Subculture Stage or capacitance, the time and the electroporation media compo cell cycle of the recipient cells may be adjusted for optimum sition. The execution of other routine adjustments will be transformation. The execution of other routine adjustments known to those of skill in the art. will be known to those of skill in the art. b. Particle Bombardment 50 c. Viral Transformation Another method for transferring a naked DNA construct i. Adenoviral Infection into cells involves particle bombardment. This method One method for delivery of the transgenic constructs depends on the ability to accelerate DNA-coated micro involves the use of an adenovirus expression vector. projectiles to a high Velocity allowing them to pierce cell Although adenovirus vectors are known to have a low membranes and enter cells without killing them (Klein et al., 55 capacity for integration into genomic DNA, this feature is 1987). The microprojectiles used have consisted of biologi counterbalanced by the high efficiency of gene transfer cally inert Substances Such as tungsten, platinum or gold afforded by these vectors. “Adenovirus expression vector” is beads. It is contemplated that in some instances DNA meant to include those constructs containing adenovirus precipitation onto metal particles would not be necessary for Sequences Sufficient to (a) Support packaging of the con DNA delivery to a recipient cell using particle bombard 60 Struct and (b) to ultimately express a transgenic construct ment. It is contemplated that particles may contain DNA that has been cloned therein. rather than be coated with DNA. Hence it is proposed that The vector comprises a genetically engineered form of DNA-coated particles may increase the level of DNA deliv adenovirus. Knowledge of the genetic organization or ery via particle bombardment but are not, in and of adenovirus, a 36 kb, linear, double-stranded DNA virus, themselves, necessary. 65 allows substitution of large pieces of adenoviral DNA with Several devices for accelerating Small particles have been foreign sequences up to 7 kb (Grunhaus and Horwitz, 1992). developed. One Such device relies on a high Voltage dis In contrast to retrovirus, the adenoviral infection of host 5,885,829 SS S6 cells does not result in chromosomal integration because as follows. A cell inoculum, resuspended in 5 ml of medium, adenoviral DNA can replicate in an episomal manner with is added to the carrier (50 ml) in a 250 ml Erlenmeyer flask out potential genotoxicity. Also, adenoviruses are structur and left Stationary, with occasional agitation, for 1 to 4 h. ally stable, and no genome rearrangement has been detected The medium is then replaced with 50 ml of fresh medium after extensive amplification. and Shaking initiated. For virus production, cells are allowed Adenovirus is particularly Suitable for use as a gene to grow to about 80% confluence, after which time the transfer vector because of its mid-sized genome, ease of medium is replaced (to 25% of the final volume) and manipulation, high titer, wide target-cell range and high adenovirus added at an MOI of 0.05. Cultures are left infectivity. Both ends of the viral genome contain 100-200 Stationary overnight, following which the Volume is base pair inverted repeats (ITRS), which are cis elements increased to 100% and shaking commenced for another 72 necessary for viral DNA replication and packaging. The h. early (E) and late (L) regions of the genome contain different Other than the requirement that the adenovirus vector be transcription units that are divided by the onset of viral DNA replication defective, or at least conditionally defective, the replication. The E1 region (E1A and E1B) encodes proteins nature of the adenovirus vector is not believed to be crucial responsible for the regulation of transcription of the viral 15 to the Successful practice of the invention. The adenovirus genome and a few cellular genes. The expression of the E2 may be of any of the 42 different known serotypes or region (E2A and E2B) results in the synthesis of the proteins Subgroups A-F. Adenovirus type 5 of Subgroup C is the for viral DNA replication. These proteins are involved in preferred Starting material in order to obtain the conditional DNA replication, late gene expression and host cell shut-off replication-defective adenovirus vector for use in the present (Renan, 1990). The products of the late genes, including the invention. This is because Adenovirus type 5 is a human majority of the viral capsid proteins, are expressed only after adenovirus about which a great deal of biochemical and Significant processing of a single primary transcript issued genetic information is known, and it has historically been by the major late promoter (MLP). The MLP, (located at used for most constructions employing adenovirus as a 16.8 m.u.) is particularly efficient during the late phase of VectOr. infection, and all the mRNA's issued from this promoter 25 AS Stated above, the typical vector according to the possess a 5'-tripartite leader (TPL) sequence which makes present invention is replication defective and will not have them preferred mRNA's for translation. an adenovirus E1 region. Thus, it will be most convenient to In a current System, recombinant adenovirus is generated introduce the transforming construct at the position from from homologous recombination between shuttle vector and which the E1-coding Sequences have been removed. provirus vector. Due to the possible recombination between However, the position of insertion of the construct within the two proviral vectors, wild-type adenovirus may be generated adenovirus Sequences is not critical to the invention. The from this process. Therefore, it is critical to isolate a single polynucleotide encoding the gene of interest may also be clone of virus from an individual plaque and examine its inserted in lieu of the deleted E3 region in E3 replacement genomic Structure. vectors as described by Karlsson et al. (1986) or in the E4 Generation and propagation of the current adenovirus 35 region where a helper cell line or helper virus complements vectors, which are replication deficient, depend on a unique the E4 defect. helper cell line, designated 293, which was transformed Adenovirus growth and manipulation is known to those of from human embryonic kidney cells by Ad5 DNA fragments skill in the art, and exhibits broad host range in vitro and in and constitutively expresses E1 proteins (Graham et al., Vivo. This group of viruses can be obtained in high titers, 1977). Since the E3 region is dispensable from the aden 40 e.g., 10°-10' plaque-forming units per ml, and they are ovirus genome (Jones and Shenk, 1978), the current aden highly infective. The life cycle of adenovirus does not ovirus vectors, with the help of 293 cells, carry foreign DNA require integration into the host cell genome. The foreign in either the E1, the D3 or both regions (Graham and Prevec, genes delivered by adenovirus vectors are episomal and, 1991). In nature, adenovirus can package approximately therefore, have low genotoxicity to host cells. No side effects 105% of the wild-type genome (Ghosh-Choudhury et al., 45 have been reported in Studies of vaccination with wild-type 1987), providing capacity for about 2 extra kb of DNA. adenovirus (Couch et al., 1963; Top et al., 1971), demon Combined with the approximately 5.5 kb of DNA that is Strating their Safety and therapeutic potential as in Vivo gene replaceable in the E1 and E3 regions, the maximum capacity transfer vectors. of the current adenovirus vector is under 7.5 kb, or about Adenovirus vectors have been used in eukaryotic gene 15% of the total length of the vector. More than 80% of the 50 expression (Levrero et al., 1991; Gomez-Foix et al., 1992) adenovirus viral genome remains in the vector backbone. and vaccine development (Grunhaus and Horwitz, 1992; Helper cell lines may be derived from human cells such Graham and Prevec, 1992). Recently, animal studies Sug as human embryonic kidney cells, muscle cells, hematopoi gested that recombinant adenovirus could be used for gene etic cells or other human embryonic mesenchymal or epi therapy (Stratford-Perricaudet and Perricaudet, 1991; thelial cells. Alternatively, the helper cells may be derived 55 Stratford-Perricaudet et al., 1990; Rich et al., 1993). Studies from the cells of other mammalian Species that are permis in administering recombinant adenovirus to different tissues Sive for human adenovirus. Such cells include, e.g., Vero include trachea instillation (Rosenfeld et al., 1991; Rosen cells or other monkey embryonic mesenchymal or epithelial feld et al., 1992), muscle injection (Ragot et al., 1993), cells. As stated above, the preferred helper cell line is 293. peripheral intravenous injections (Herz and Gerard, 1993) Recently, Racher et al. (1995) disclosed improved meth 60 and Stereotactic inoculation into the brain (Le Gal La Salle ods for culturing 293 cells and propagating adenovirus. In et al., 1993). one format, natural cell aggregates are grown by inoculating ii. AAV Infection individual cells into 1 liter Siliconized spinner flaskS Adeno-associated virus (AAV) is an attractive vector (Techne, Cambridge, UK) containing 100-200 ml of System for use in the present invention as it has a high medium. Following stirring at 40 rpm, the cell viability is 65 frequency of integration and it can infect nondividing cells, estimated with trypan blue. In another format, Fibra-Cel thus making it useful for delivery of genes into mammalian microcarriers (Bibby Sterlin, Stone, UK) (5 g/l) is employed cells in tissue culture (Muzyczka, 1992). AAV has a broad 5,885,829 57 58 host range for infectivity (Tratschin, et al., 1984; Laughlin, Two long terminal repeat (LTR) sequences are present at the et al., 1986; Lebkowski, et al., 1988; McLaughlin, et al., 5' and 3' ends of the viral genome. These contain Strong 1988), which means it is applicable for use with the present promoter and enhancer Sequences and are also required for invention. Details concerning the generation and use of integration in the host cell genome (Coffin, 1990). rAAV vectors are described in U.S. Pat. No. 5,139,941 and In order to construct a retroviral vector, a nucleic acid U.S. Pat. No. 4,797.368, each incorporated herein by refer encoding a transgene of interest is inserted into the viral genome in the place of certain Viral Sequences to produce a CCC. Virus that is replication-defective. In order to produce Studies demonstrating the use of AAV in gene delivery Virions, a packaging cell line containing the gag, pol, and include LaFace et al. (1988); Zhou et al. (1993); Flotte et al. env genes but without the LTR and packaging components (1993); and Walsh et al. (1994). Recombinant AAV vectors is constructed (Mann et al., 1983). When a recombinant have been used Successfully for in Vitro and in Vivo trans plasmid containing a cDNA, together with the retroviral duction of marker genes (Kaplitt, et al., 1994, Lebkowski, et LTR and packaging Sequences is introduced into this cell al., 1988; Samulski, et al., 1989; Shelling and Smith, 1994; line (by calcium phosphate precipitation for example), the Yoder, et al., 1994, Zhou, et al., 1994; Hermonat and packaging Sequence allows the RNA transcript of the recom Muzyczka, 1984; Tratschin, et al., 1985; McLaughlin, et al., 15 binant plasmid to be packaged into Viral particles, which are 1988) and genes involved in human diseases (Flotte, et al., then Secreted into the culture media (Nicolas and 1992; Luo, et al., 1994; Ohi, et al., 1990; Walsh, et al., 1994; Rubenstein, 1988; Temin, 1986; Mann et al., 1983). The Wei, et al., 1994). Recently, an AAV vector has been media containing the recombinant retroviruses is then approved for phase I human trials for the treatment of cystic collected, optionally concentrated, and used for gene trans fibrosis. fer. Retroviral vectors are able to infect a broad variety of AAV is a dependent parvovirus in that it requires coin cell types. However, integration and Stable expression fection with another virus (either adenovirus or a member of require the division of host cells (Paskind et al., 1975). the herpesvirus family) to undergo a productive infection in Concern with the use of defective retrovirus vectors is the cultured cells (Muzyczka, 1992). In the absence of coinfec potential appearance of wild-type replication-competent tion with helper virus, the wild type AAV genome integrates 25 Virus in the packaging cells. This can result from recombi through its ends into human chromosome 19 where it resides nation events in which the intact Sequence from the recom in a latent state as a provirus (Kotin et al., 1990; Samulski binant virus inserts upstream from the gag, pol, env et al., 1991). raAV, however, is not restricted to chromo Sequence integrated in the host cell genome. However, new Some 19 for integration unless the AAV protein is also packaging cell lines are now available that should greatly expressed (Shelling and Smith, 1994). When a cell carrying decrease the likelihood of recombination (Markowitz et al., an AAV provirus is Superinfected with a helper virus, the 1988; Hersdorffer et al., 1990). AAV genome is “rescued' from the chromosome or from a iv. Other Viral Vectors recombinant plasmid, and a normal productive infection is Other viral vectors may be employed as constructs in the established (Samulski, et al., 1989; McLaughlin, et al., 1988; present invention. Vectors derived from Viruses Such as Kotin, et al., 1990; Muzyczka, 1992). 35 vaccinia virus (Ridgeway, 1988; Baichwal and Sugden, Typically, recombinant AAV (ra AV) virus is made by 1986; Coupar et al., 1988) and herpesviruses may be cotransfecting a plasmid containing the gene of interest employed. They offer several attractive features for various flanked by the two AAV terminal repeats (McLaughlin et al., mammalian cells (Friedmann, 1989; Ridgeway, 1988; 1988; Samulski et al., 1989; each incorporated herein by Baichwal and Sugden, 1986; Coupar et al., 1988; Horwich reference) and an expression plasmid containing the wild 40 et al., 1990). type AAV coding Sequences without the terminal repeats, for With the recent recognition of defective hepatitis B example pIM45 (McCarty et al., 1991; incorporated herein Viruses, new insight was gained into the Structure-function by reference). The cells are also infected or transfected with relationship of different viral Sequences. In vitro Studies adenovirus or plasmids carrying the adenovirus genes showed that the virus could retain the ability for helper required for AAV helper function. raAV virus stocks made 45 dependent packaging and reverse transcription despite the in Such fashion are contaminated with adenovirus which deletion of up to 80% of its genome (Horwich et al., 1990). must be physically separated from the rAAV particles (for This Suggested that large portions of the genome could be example, by cesium chloride density centrifugation). replaced with foreign genetic material. Chang et al. recently Alternatively, adenovirus vectors containing the AAV cod introduced the chloramphenicol acetyltransferase (CAT) ing regions or cell lines containing the AAV coding regions 50 gene into duck hepatitis B virus genome in the place of the and Some or all of the adenovirus helper genes could be used polymerase, Surface, and pre-Surface coding Sequences. It (Yang et al., 1994a, Clark et al., 1995). Cell lines carrying was cotransfected with wild-type virus into an avian the rAAV DNA as an integrated provirus can also be used hepatoma cell line. Culture media containing high titers of (Flotte et al., 1995). the recombinant virus were used to infect primary duckling iii. Retroviral Infection 55 hepatocytes. Stable CAT gene expression was detected for at The retroviruses are a group of Single-Stranded RNA least 24 days after transfection (Chang et al., 1991). viruses characterized by an ability to convert their RNA to In still further embodiments of the present invention, the double-stranded DNA in infected cells by a process of nucleic acids to be delivered are housed within an infective reverse-transcription (Coffin, 1990). The resulting DNA then Virus that has been engineered to express a specific binding Stably integrates into cellular chromosomes as a provirus 60 ligand. The virus particle will thus bind specifically to the and directs Synthesis of viral proteins. The integration results cognate receptors of the target cell and deliver the contents in the retention of the viral gene Sequences in the recipient to the cell. A novel approach designed to allow specific cell and its descendants. The retroviral genome contains targeting of retrovirus vectors was recently developed based three genes, gag, pol, and env that code for capsid proteins, on the chemical modification of a retrovirus by the chemical polymerase enzyme, and envelope components, respec 65 addition of lactose residues to the viral envelope. This tively. A sequence found upstream from the gag gene modification can permit the Specific infection of hepatocytes contains a signal for packaging of the genome into Virions. Via Sialoglycoprotein receptorS. 5,885,829 59 60 Another approach to targeting of recombinant retroviruses Ovirus assisted transfection. Increased transfection efficien was designed in which biotinylated antibodies against a cies have been reported in cell Systems using adenovirus retroviral envelope protein and against a Specific cell recep coupled systems (Kelleher and Vos, 1994; Cotten et al., tor were used. The antibodies were coupled via the biotin 1992; Curiel, 1994), and the inventors contemplate using the components by using Streptavidin (Roux et al., 1989). Using Same technique to increase transfection efficiencies. antibodies against major histocompatibility complex class I h. Receptor Mediated Transfection and class II antigens, they demonstrated the infection of a Still further constructs that may be employed to deliver variety of human cells that bore those Surface antigens with the transgenic construct to the target cells are receptor an ecotropic virus in vitro (Roux et al., 1989). d. Calcium Phosphate Co-Precipitation or DEAE mediated delivery vehicles. These take advantage of the Dextran Selective uptake of macromolecules by receptor-mediated In other preferred embodiments of the present invention, endocytosis that will be occurring in the target cells. In view the transgenic construct is introduced to the cells using of the cell type-specific distribution of various receptors, this calcium phosphate co-precipitation. Mouse primordial germ delivery method adds a degree of Specificity to the present cells have been transfected with the SV40 large T antigen, invention. Specific delivery in the context of another mam with excellent results (Watanabe et al., 1997). Human KB 15 malian cell type is described by Wu and Wu (1993; incor cells have been transfected with adenovirus 5 DNA (Graham porated herein by reference). and Van Der Eb, 1973) using this technique. Also in this Certain transgenic delivery constructs comprise a cell manner, mouse L(A9), mouse C127, CHO, CV-1, BHK, receptor-specific ligand and a DNA-binding agent. Others NIH3T3 and HeLa cells were transfected with a neomycin comprise a cell receptor-specific ligand to which the DNA marker gene (Chen and Okayama, 1987), and rat hepato construct to be delivered has been operatively attached. cytes were transfected with a variety of marker genes (Rippe Several ligands have been used for receptor-mediated gene et al., 1990). transfer (Wu and Wu, 1987; Wagner et al., 1990; Ferkol et In another embodiment, the expression construct is deliv al., 1993; Perales et al., 1994; Myers, EPO 0273085), which ered into the cell using DEAE-dextran followed by poly establishes the operability of the technique. ethylene glycol. In this manner, reporter plasmids were 25 In other embodiments, the DNA delivery vehicle compo introduced into mouse myeloma and erythroleukemia cells nent may comprise a specific binding ligand in combination (Gopal, 1985). with a liposome. The nucleic acids to be delivered are e. Direct Microinjection or Sonication Loading housed within the liposome and the Specific binding ligand Further embodiments of the present invention include the is functionally incorporated into the liposome membrane. introduction of the transgenic construct by direct microin The liposome will thus specifically bind to the receptors of jection or Sonication loading. Direct microinjection has been the target cell and deliver the contents to the cell. Such used to introduce nucleic acid constructs into Xenopus Systems have been shown to be functional using Systems in oocytes (Harland and Weintraub, 1985), and LTK fibroblasts which, for example, epidermal growth factor (EGF) is used have been transfected with the thymidine kinase gene by in the receptor-mediated delivery of a nucleic acid to cells Sonication loading (Fechheimer et al., 1987). 35 that exhibit upregulation of the EGF receptor. f. Liposome Mediated Transformation In still further embodiments, the DNA delivery vehicle In a further embodiment of the invention, the transgenic component of the delivery vehicles may be a liposome itself, construct may be entrapped in a liposome. Liposomes are which will preferably comprise one or more lipids or gly vesicular structures characterized by a phospholipid bilayer coproteins that direct cell-specific binding. For example, membrane and an inner aqueous medium. Multilamellar 40 Nicolau et al. (1987) employed lactosyl-ceramide, a liposomes have multiple lipid layerS Separated by aqueous galactose-terminalasialoganglioside, incorporated into lipo medium. They form Spontaneously when phospholipids are Somes and observed an increase in the uptake of the insulin Suspended in an excess of aqueous Solution. The lipid gene by hepatocytes. It is contemplated that the transgenic components undergo Self-rearrangement before the forma constructs of the present invention can be specifically deliv tion of closed Structures and entrap water and dissolved 45 ered into the target cells in a similar manner. solutes between the lipid bilayers (Ghosh and Bachhawat, 6. Homologous Recombination 1991). Also contemplated is a transgenic construct com Although genetic transformation tends to be quite plexed with Lipofectamine (Gibco BRL). efficient, it is also accompanied by problems associated with Liposome-mediated nucleic acid delivery and expression random insertion. Random integration can lead to the inac of foreign DNA in vitro has been very successful (Nicolau 50 tivation of essential genes, or to the abberant expression of and Sene, 1982; Fraley et al., 1979; Nicolau et al., 1987). the introduced gene. Additional problems associated with Wong et al. (1980) demonstrated the feasibility of liposome genetic transformation include mosaicism due to multiple mediated delivery and expression of foreign DNA in cul integrations, and technical difficulties associated with gen tured chick embryo, HeLa and hepatoma cells. eration of replication defective recombinant Viral vectors. In certain embodiments of the invention, the lipoSome 55 Some of these drawbacks can be overcome by the utili may be complexed with a hemagglutinating virus (HVJ). Zation of a technique known as homologous recombination This has been shown to facilitate fusion with the cell (Koller and Smithies, 1992). This technique allows the membrane and promote cell entry of liposome-encapsulated precise modification of existing genes, overcomes the prob DNA (Kaneda et al., 1989). In other embodiments, the lems of positional effects and insertional inactivation, and lipoSome may be complexed or employed in conjunction 60 allows the inactivation of Specific genes, as well as the with nuclear non-histone chromosomal proteins (HMG-1) replacement of one gene for another. Methods for homolo (Kato et al., 1991). In yet further embodiments, the liposome gous recombination are described in U.S. Pat. No. 5,614, may be complexed or employed in conjunction with both 396, incorporated herein in its entirety by reference. HVJ and HMG-1. Thus a preferred method for the delivery of transgenic g. Adenoviral ASSisted Transfection 65 constructs involves the use of homologous recombination. In certain embodiments of the present invention, the Homologous recombination relies, like antisense, on the transgenic construct is introduced into the cell using aden tendency of nucleic acids to base pair with complementary 5,885,829 61 62 Sequences. In this instance, the base pairing Serves to mercial Sale Such as, e.g., injection or blow-molded plastic facilitate the interaction of two Separate nucleic acid mol containers into which the desired vials are retained. Irre ecules So that Strand breakage and repair can take place. In Spective of the number or type of containers, the kits of the other words, the “homologous” aspect of the method relies invention are typically packaged with instructions for use of on Sequence homology to bring two complementary the kit components. Sequences into close proximity, while the “recombination' The following examples are included to demonstrate aspect provides for one complementary Sequence to replace preferred embodiments of the invention. It should be appre the other by virtue of the breaking of certain bonds and the ciated by those of skill in the art that the techniques formation of others. disclosed in the examples that follow represent techniques Put into practice, homologous recombination is used as discovered by the inventors to function well in the practice follows. First, a site for integration is selected within the host of the invention, and thus can be considered to constitute cell. Sequences homologous to the integration site are then preferred modes for its practice. However, those of skill in included in a genetic construct, flanking the Selected gene to the art should, in light of the present disclosure, appreciate be integrated into the genome. Flanking, in this context, that many changes can be made in the Specific embodiments Simply means that target homologous Sequences are located 15 that are disclosed and Still obtain a like or Similar result both upstream (5') and downstream (3') of the selected gene. without departing from the Spirit and Scope of the invention. These Sequences should correspond to Some Sequences upstream and downstream of the target gene. The construct EXAMPLE I is then introduced into the cell, thus permitting recombina tion between the cellular Sequences and the construct. Isolation and Expansion of Pulp-derived Fibroblasts AS a practical matter, the genetic construct will normally Cells were explanted and propagated from adult human act as far more than a vehicle to insert the gene into the dental pulps, as described by Rutherford et. al. (1992a). genome. For example, it is important to be able to Select for Teeth are Sectioned into coronal and root fragments and the recombinants and, therefore, it is common to include within pulps extirpated with a Spoon excavator. This is best done the construct a Selectable marker gene. This gene permits 25 within 24 hours of extraction. If the pulp tissues cannot be Selection of cells that have integrated the construct into their harvested immediately, it is best to store the teeth at 4 C. genomic DNA by conferring resistance to various biostatic submerged in DMEM supplemented with penicillin, strep and biocidal drugs. In addition, this technique may be used tomycin and neomycin at three times normal concentration to “knock-Out' (delete) or interrupt a particular gene. This is (3x). accomplished by including a mutated or vastly deleted form The tissues are suspended in DMEM, 10% fetal bovine of the heterologous gene between the flanking regions Serum, Supplemented with penicillin, Streptomycin, and neo within the construct. The arrangement of a construct to effect mycin (3x) and minced to pieces approximately 1–2 mm, homologous recombination might be as follows: and placed into 25 ml tissue culture flasks. When sufficient numbers of cells have migrated from the cultured tissue 35 ... vectors'-flanking sequence'selected geneselectable marker pieces (explants) and proliferated So as to nearly cover the gene flanking sequence-3'-vector . . . Surface of the flask, the cultures are harvested, diluted, and placed into more vessels or concentrated and cryopreserved. Thus, using this kind of construct, it is possible, in a Single Cells were then cultured in DMEM (GIBCO; Grand recombinatorial event, to (i) “knock out an endogenous Island, N.Y.) Supplemented with penicillin/streptomycin gene, (ii) provide a Selectable marker for identifying Such an 40 event and (iii) introduce a transgene for expression. (GIBCO), and 10% fetal calfserum (Hyclone; Logan, Utah). Another refinement of the homologous recombination Standard cell culture methods were employed. Cells approach involves the use of a “negative' Selectable marker. between passages 5 to 8 were used in all Studies. One example of the use of the cytosine deaminase gene in EXAMPLE II a negative selection method is described in U.S. Pat. No. 45 5,624,830. The negative selection marker, unlike the select Cell Seeding and Culture on Synthetic Matrices able marker, causes death of cells which express the marker. Thus, it is used to identify undesirable recombination events. Cells were seeded onto a non-woven matrix (thickness=3 When Seeking to Select homologous recombinants using a mm, bulk density=50 mg/cc) formed from polyglycolic acid Selectable marker, it is difficult in the initial Screening Step 50 (PGA) fibers (12 um in diameter) (Albany Intl.; Taunton to identify proper homologous recombinants from recom Mass.). binants generated from random, non-Sequence Specific The PGA fibers were prepared by extrusion of molten events. These recombinants also may contain the Selectable polymer, which was Subsequently drawn out and annealed to marker gene and may express the heterologous protein of maximize the crystallinity. The fibers were then cut, typi interest, but will, in all likelihood, not have the desired 55 cally into 2 inch lengths, and the cut fibers were crimped. phenotype. By attaching a negative Selectable marker to the The crimped fibers were then assembled into a mat with the construct, but outside of the flanking regions, one can Select fibers randomly arranged. Fiber mats were then “needled', against many random recombination events that will incor using a board of needles So that the fibers became entangled. porate the negative Selectable marker. Homologous recom The needled mats were then pressed with a heated platen to bination should not introduce the negative Selectable marker, 60 produce a product with the desired thickneSS and bulk as it is outside of the flanking Sequences. density. H. Kits Matrices with desired dimensions are then cut and Ster All the essential materials and reagents required for the ilized prior to use. A variety of Sterilization techniques are various aspects of the present invention may be assembled possible, including Y-radiation and the use of ethylene oxide. together in a kit. The kits of the present invention also will 65 Cells, confluent in tissue cultured flasks, were rinsed with typically include a means for containing the Vials compris phosphate buffered saline, and incubated with 0.05% ing the desired components in close confinement for com trypsin/0.5 mM EDTA to remove adherent cells. Cells from 5,885,829 63 64 multiple flasks were pooled, and the cells concentrated to 3). Calculation of the cellular density in these tissues over 5x10 cells/ml in the tissue culture medium. Each PGA time revealed an increase of almost two orders of magnitude Scaffold (1x1 cm Square) was placed in a well of a 6-well to a final density of 4.6x10" cells/ml at 60 days (FIG. 4). dish (Fisher Sci.), and seeded with 0.25 ml of the cell The mean cellularity calculated from histological croSS Suspension. sections of 3 freshly extracted pulp tissues was 1500+400 Seeded scaffolds were placed in an incubator (5% CO, cells/mm, while the engineered pulp tissues had a mean 37° C) for 30 minutes to allow cell adhesion, and 2 ml of cellularity of 330+50 and 2700+1100 cells/mm after 28 and tissue cultured medium was then added to each Scaffold. 60 days, respectively, in culture. Thus, the cellularity of the Alternatively, 1x10 cells/ml in 5 ml of tissue culture engineered tissues was in the same range as the equivalent medium was incubated with the scaffold for 24 hours by adult tissue. stirring in a spinner flask at 20-30 rpm. Scaffolds were Photomicrographs of histological Sections of engineered cultured for time periods ranging from 1 to 60 dayS. pulp tissue Stained with hematoxylin and eosin showed a grOSS Similarity to normal adult pulp tissue. EXAMPLE III 15 EXAMPLE IV TiSSue Regeneration A. Analytical Methods Biodegradable Polymer Matrices GroSS measurements were made of the cell-polymer con Synthetic, biodegradable polymers are used as they are Structs at each time point. Samples were prepared for attractive Scaffold materials that can be readily produced Visualization with a Scanning electron microScope by fixing with a wide range of reproducible properties and Structures. with a 1% Solution of glutaraldehyde, dehydrating in A. Structural Materials methanol, and Sputter coated (Desk II; Denton Vacuum, 1. PGA and PLA Cherry Hill, N.J.) with gold. Photomicrographs were taken Aliphatic polyesters of the poly(C.-hydroxy acids) have on Polaroid TM 55 film. Samples were prepared for sectioning 25 the general formula --O-CH(R)-CO-- which and staining by fixing with a solution of 3.7% formalin in derive from corresponding HO-CH(R)-COOH where phosphate buffered Saline. Thin Sections (5 um) were cut and R=H in the case of glycolic acid (GA) and R=CH3 in the Stained with hematoxylin and eosin using Standard tech case of lactic acid (LA), the latter being chiral, i.e., D- or niques. L-isomer is possible. These polymerS have been used in The density of cells in histological Sections of cell bone osteosynthesis and reconstruction (Vert et al., 1984) polymer constructs and native pulp tissue (adult third and in drug delivery (Gombotz and Pettit, 1995). molars) was determined by counting the number of cellular a. Synthesis nuclei per area. Photomicrographs were taken with Kodak PGA and PLA can be prepared by two different routes, GoldTM film. namely, polycondensation and ring opening polymerization The DNA content of cell-polymer constructs was deter 35 (ROP). Generally, the simple polycondensation is less mined using a variation of a dye binding assay (Kim et al., expensive, but the resulting polymers have low and uncon 1988). In brief, cell-polymer constructs were flash frozen in trolled molecular weight and it is difficult to prepare copoly liquid nitrogen, lyophilized, and stored at -70° C. The mers (Gilding and Reed, 1979, Kricheldorf and Kreiser Samples were Subsequently digested in a Solution of 0.5 Saunders, 1996). It is believed that the antimony trioxide mg/ml Proteinase K (Sigma; St. Louis Mo.), 1% SDS, 50 40 catalyst typically used to effect polycondensation acts as mM Tris-HCl, 0.1M EDTA, 0.2M NaCl at a pH of 9.0. The both polymerizing and depolymerizing agent. Moreover, digestion was carried out at 55 C. for 6 hr with occasional glycolic and lactic acids have a great tendency to gentle agitation. The DNA content in digested Samples was cyclodimerize under these conditions and this renders determined by assaying the binding of a dye, Hoechst Simple polycondensation an unsuitable method. 33258, that specifically binds DNA (Kim et al., 1988) with 45 The preferred method for producing high molecular a fluorimeter (DynaQuant 2000; Hoefer Sci. Inst.; San weight polymers is ROP of the cyclic dimer, glycolide Francisco Calif.). Calibration of the fluorimeter was per (and/or lactide). Depending on the catalyst involved, three formed with known concentrations of calf thymus DNA different mechanisms have been reported: cationic, anionic (Hoefer Sci. Inst.), and a calibration curve was constructed and insertion. Among these, the insertion mechanism using for this assay using known numbers of cultured cells. 50 metal alkoxides or carboxylates is the most desirable path Cell densities were determined by dividing the number of way and is the choice in commercial production (Frazza and cells in each construct (determined with DNA assay) by the Schmitt, 1971). Typical examples of catalysts of this class Volume of the tissue (determined with gross measurement). are aluminum, zinc, titanium, Zirconium, antimony, tin(IV) B. Results and tin(II) alkoxides or carboxylates. 55 The insertion mechanism allows the preparation of high The polymer scaffolds utilized as synthetic extracellular molecular weight polylactides without racemization up to matrices were comprised of a non-woven array of polygly temperatures above 150° C. (Kricheldorf and Kreiser colic acid fibers (FIG. 1). Saunders, 1996). The mechanism of ROP of lactones has Pulp-derived fibroblasts adhered to the polyglycolic acid been reviewed by Penczek and Slomkowski (1989). The tin fibers of the polymer mesh following cell Seeding, spread 60 catalyst, tin(II) octoate, was used extensively because of its extensively over the fibers and spanned the polymer fibers acceptance by the FDA as a food stabilizer. (FIG. 2). Quantitation of the number of adherent cells after The polymerization of glycolide can be carried out in bulk one day in culture revealed that 2.6+0.8x10 cells were at 220 C. for 4 h, at which time a 96% conversion and present in each Scaffold. This corresponds to an initial cell molecular weights from 10" to 10° have been reported. density of approximately 9x10 cells/ml. 65 Copolymerization of glycolide with lactide has also been The cells proliferated on the scaffolds over time, and investigated. The reactivity ratios at 200 C. have been ultimately occupied the spaces between polymer fibers (FIG. found to be 2.8 for glycolide and 0.2 for lactide. This 5,885,829 65 66 indicates that copolymers of glycolic and lactic acids will PLA is employed much more often than d-PLA, since the have broad compositional ranges, with glycolide always hydrolysis of PLA yields L-lactic acid which is the naturally being preferentially polymerized at low conversions and occurring Stereoisomer of lactic acid. Whereas PLA pos lactide being incorporated to ever-increasing extents as the sesses about 37% crystallinity, the optically inactive poly glycolide is depleted (Gilding and Reed, 1979). 5 (DL-lactic acid) (dl-PLA) is amorphous. The difference in During the advanced stages of most ROPs, additional the crystallinity of dl-PLA and PLA has important practical reactions Such as ester-ester interchange and chain unzip outgrowths. For instance, the amorphous dl-PLA is usually ping may take place. The extents of these reactions are considered in drug delivery application where a homoge affected by the reactivity of the ester moieties. These events can have a significant effect on the composition of the final neous dispersion of the active Species within a monophasic product. Due to the ester eXchange, cyclic dimer, trimer and, matrix is desired (Engelberg and Kohn, 1991). However, the to a less extent, cyclic oligomers could be found along with Semicrystalline PLA is preferred in case where high the reformed monomer. In the case of copolymerization, mechanical Strength and toughneSS are required, for additional randomization of the polymer chain would occur example, in orthopedic devices (Leenslag et al., 1987, as a consequence of the ester-ester exchange of different Vainionpaa et al., 1987; Hay et al., 1988). It is pertinent to ester moieties (Shalaby and Johnson, 1994). The microstruc 15 note that Y-irradiation of PLA causes chain Scission, tures of PLGA copolymers can be determined by both crosslinking and a decrease in crystallinity (Gupta and proton and carbon NMR spectroscopy (Ksaperczyk, 1996). Deshmuth, 1983). Therefore, caution should be taken when It has been reported that the block lengths increase and, at Sterilizing the polymer matrices by Y-irradiation. the same time, the extent of transesterification decreases To widen the range of materials properties exhibited by with decreasing polymerization temperature. PGA, copolymers of GA and LA (PLGA) have been studied. b. Properties of PGA and PLA Tissue Engineering Whereas PGA is highly crystalline, PLGA usually exhibit (1) Mechanical Strength and Morphology lower crystallinity and Tm (Gilding and Reed, 1979). For PGA was first developed as the synthetic absorbable example, while PGA and PLA are partially crystalline, 50:50 suture, Dexon (Frazza and Schmitt, 1971). PGA has high PLGA is entirely amorphous. These morphological changes crystallinility, a high melting point and low Solubility in 25 result in an increase in the rates of hydration and hydrolysis. organic solvents (Table 7). The polymer (fiber grade, inher Thus, copolymers tend to degrade more rapidly than PGA ent Viscosity=1.2-1.6 dL/g in hexafluoro-isopropanol) can and PLA (Mooney et al., 1995b). be spun into multifilament or monofilament for the (2) Biodegradation production of braided and monofilament Sutures, respec The degradation mechanism of PGA and copolymers in tively (Frazza and Schmitt, 1971; Chujo et al., 1967). A vitro is usually regarded as bulk erosion (Gombotz and typical suture braid has a tensile strength of 80-100 Kipsi Pettit, 1995). This is evident from the fact that a significant (Table 8). Owing to the hydrophilic nature of PGA, Dexon molecular weight decrease usually precedes monomer Sutures tend to lose their mechanical strength rapidly (50%) release from the polymer Samples. This mechanism of over a period of 2 weeks and are absorbed in about 4 weeks degradation may be undesirable in certain applications. The after implantation (Frazza and Schmitt, 1971; Reed and 35 relatively rapid release of large quantities of acid (glycolic Gilding, 1981; Katz and Turner, 1970). and/or lactic acids) may lead to a local acidosis if a large mass of these polymerS is present in a concentrated form TABLE 7 (e.g., a Solid pin). However, highly porous scaffolds are typically utilized in tissue engineering applications, and Crystallinity and Thermal Properties of PGA, PLA and Copolymers 40 contain a relatively low mass of polymer per unit volume The highly porous Structure of the Scaffolds assists cell % Crystallinity Tim Tg penetration as well as polymer degradation (Mooney et al., PGA 46-52 225 36 1994b; 1995a; 1995b). The rate of degradation will be 90:10 PGLA 40 210 37 SO:SOPGLA O None 55 affected by the morphology of the scaffold and the large 45 PLA 37 185 57 Surface areas Speed up the diffusion of water molecules into d-PLA O None N/A the bulk of the polymers when they are placed in an aqueous environment (e.g., in vivo). Adapted from Wong and Mooney, 1997. The polymers undergo random chain Scission by Simple hydrolysis of the ester bond linkage and the monomer 50 diffuses out of the polymer bulk into water (Reed and TABLE 8 Gilding, 1981). It is important to note that loss of mechanical Mechanical Properties of PGA and 90:10 PGLA strength of PGA is faster when the polymer is incubated at a temperature higher than its Tg. This indicates that the PGA (Dexon) 90:10 PGLA (Vicryl) glassy State protects PGA from hydrolysis Since all short Tensile strength (Kpsi) 106 95 55 term chain motions are frozen. Water diffusion, and there Knot strength (Kpsi) 65 63 fore hydrolysis, is more facile at temperatures above Tg. It Elongation (%) 24 25 is also relevant to mention that the Tgs of PGA and some copolymers are very close to the physiological temperature. Adapted from Wong and Mooney, 1997. Polymeric materials may undergo Significant Structural The presence of an extra methyl group poly(L-lactic acid) 60 change after implantation due to water penetration and loSS (PLA) or poly(D-lactic acid) (d-PLA) makes them more of mechanical Strength. It is also speculated that enzymatic hydrophobic than PGA. For instance, films of PLA only take action may partially contribute to biodegradation of PGA in up approximately 2% water (Gilding and Reed, 1979). In WVO. addition, the ester bond in PLA is less labile to hydrolysis The chemical compositions and the ratio of monomers due to steric hindrance of the methyl group. Therefore, PLA 65 used in the polymerization reaction Strongly influence the degrades much slower than PGA (Reed and Gilding, 1981) degradation characteristics of the copolymer. The degrada and has higher Solubility in organic Solvents. tion rates for copolymers of GA and LA have been shown to 5,885,829 67 68 be influenced by factors which affect polymer chain packing, b. Other Polyesters i.e., crystallinity, and hydrophobicity. Since degradation is Properties of polyesters can also be varied by changing induced by hydrolysis, a crystalline Structure or hydrophobic the Structures of the polymer backbones. Polycaprolactone polymer composition disfavors dissolution and degradation. (PCL), having two more carbon atoms than PGA on the Gombotz and Pettit (1995) summarized the specific fac polymer backbone, has been Studied as a Substrate for tors affecting copolymer crystallinity and hydrophobicity: biodegradation and as a matrix for drug release Systems (i) the ratio of lactide to glycolide monomer in the copolymer, (ii) the Stereoregularity of the monomer units in (Huang, 1989). Its degradation in vivo is much slower than the polymer affects polymer chain packing, (iii) randomness PGA, therefore, it is suitable for controlled release devices of lactide and glycolide decrease the ability of chains to with long in vivo life times (1–2 years). PCL can be prepared crystallize, and (iv) low molecular weight polymers degrade by anionic ring opening polymerization of e-caprolactone faster than high molecular weight polymers, especially when using metal hydroxide initiators (Jerome and Teyssie, 1989). the end groups are free acid rather than capped with ester or Poly-B-hydroxy acid can be prepared by both cationic and other groups. Mass loSS from polymer Samples comprised of anionic ring opening polymerizations (Penczek and PLA is insignificant in the experimental time period (~50 Slomkowski, 1989; Jerome and Teyssie, 1989). For weeks). However, those comprised of copolymers of GA and 15 example, 100% syndiotactic poly(B-DL-hydroxybutyrate) LA or d1-PLA degrade much faster (the higher the glycolic has been prepared by treating the corresponding lactone with acid content, the higher the degradation rate) (Mooney et al., cyclic dibutyltin initiators to yield high molecular weight 1995b). polymers (Kricheldorf and Lee, 1995). Bacteria also pro The presence of monomers and low molecular weight duce the chiral, isotactic poly(B-D-hydroxybutyrate) as a cyclic oligomers in absorbable polymers should be avoided, highly crystalline biopolymer (Holmes, 1988). for they degrade much more rapidly than the polymers and Numerous analogs of poly(3-hydroxy acid) have been can lead to undesirable chemical and biological effects. Synthesized either chemically by the ring opening polymer (Shalaby and Johnson, 1994) It has been shown that poly ization or biologically by feeding unusual carbon Sources to lactide with increased monomer content exhibits a higher bacteria (Timmins and Lenz, 1994). The microbial synthesis rate of bioabsorption and a more drastic decrease of molecu 25 of polyesters has been reviewed by Gross (1994). Due to lar weight. (Nakamura et al., 1989) their biocompatibility and biodegradability, different blends 2. Other Chemistries for Tissue Engineering of polycaprolactone, poly(3-hydroxybutyrate) and other a. Chemical Modifications of PGA/PLA polymers have been fabricated for medical devices (Yasin Little modification of these polymerS is possible because and Tighe 1992), drug delivery applications (Wang, 1989) there are no other functional groups on the Side chain, except and cell microencapsulation (Giunchedi et al., 1994; Emb the methyl of the lactic acid residue. One possibility to leton and Tighe, 1993). modify the properties of these polymerS is to form copoly Surface-eroding polymer matrices are attractive for a mers with residues having more diverse side chain Variety of tissue engineering applications. The monomer Structures, e.g., lysine. release would be steady over the lifetime of the matrices in A new monomer, 3-(Ne-benzoxycarbonyl-L-lysyl)-6-L- 35 contrast to PLA and PGA. In addition, the gradual loss of methyl-2,5-morpholinedione, was bulk copolymerized with polymer from the surface of the scaffold may allow the L,L-lactide in the presence of Stannous Octoate as catalyst Surrounding tissue to Serially fill the Space vacated by the using the same ROP techniques utilized for lactide and polymer. glycolide (Barrera et al., 1993). The lysine content was Polyorthoesters are an example of Surface-eroding poly determined by NMR technique to be approximately 1.3 40 mers. The hydrophobic character of the polymer limits water mole %. penetration and hydrolysis to only the exterior Surface of the A poly(lactide-co-lysine) functionalized with peptide con polymer matrix (Heller, 1985). Thus the surface erosion is taining the arginine-glycine-aspartate (RGD) sequence was much faster than that of the bulk. The chemical and physical prepared by removal of the benzyOxycarbonyl protecting properties of polyorthoesters have been reviewed (Heller group on the lysyl residue and peptide coupling. The peptide 45 and Daniels, 1994) and depend on the chemical structures of concentration was found to be approximately 3.1 mmol/g the constituent monomers. For example, reaction of bis which could be translated into a peptide Surface density of (ketene acetal) with rigid trans-cyclohexane dimethanol 310 fmol/cm. A surface density of as low as 1 fmol/cm of produce a rigid polymer with a Tg of 110° C., whereas that an RGD peptide has been previously determined to promote of the flexible diol 1,6-hexanediol produces a soft material cell adhesion to an otherwise nonadherent Surface (Massia 50 having a Tg of 20° C. Mixture of the two diol results in and Hubbell, 1991). Therefore, by carefully processing the polymers having intermediate Tg. copolymer, biodegradable films with cell adhering proper Degradation of the polymerS is acid-induced, and degra ties can be prepared from the copolymer of lactide and dation rates can be increased by adding acidic excipients or lysine. by increasing the hydrophilicity of the polymer matrix. Other Strategies have also been employed to widen the 55 Conversely, degradation can be retarded by using basic properties of polylactides. For example, PLA has also been excipients such as Mg(OH) (Gombotz and Pettit, 1995). Synthesized as an acrylic macromonomer and Subsequently Current applications of these polymers include Sustained copolymerized with polar acrylic monomers (e.g., drug delivery as well as hard and soft tissue fixation (Heller 2-hydroxyethylmethacrylate) (Barakat et al., 1996). These and Daniels, 1994). polymers were Studied as amphiphilic graft copolymers for 60 Poly orthoformate, polycarbonate, poly(oxyethylene drug delivery purposes. The Surface properties of these glycolate), poly(1,4-butylene diglycolate) and polymers may be controlled by the ratio of the PLA graft are other biodegradable polymers that may have applications length and copolymer content, and can be potentially used to in tissue engineering. Many of these polymers have been control the drug release profile and biodistribution. Other previously been utilized as drug delivery matrices (Huang, examples of this approach include grafting PLA blocks to 65 1989). geraniol and pregnenolone (Kricheldorf and Kreiser There are therefore a large number of polyesters and Saunders, 1996). analogs that are biodegradable. Their mechanical properties 5,885,829 69 70 can be controlled largely by the chemical Structures of the Sequences glycine-any amino acid-glycine-Valine-proline constituent building blocks and can be varied from tough to (GXGVP). The polymers were synthesized by the self elastic. The biocompatibility of these polymerS is presumed condensation of the activated p-nitrophenol ester of the to result from non-toxic degradation products. Bioactive pentapeptide building blocks (Prasad et al., 1985). The elements can be attached to this class of materials in order molecular weight of these polymers are considered to be to mimic natural extracellular matrix molecules. higher than 50 000. The polymers can be cross-linked by c. Polypeptides Y-irradiation to form an insoluble matrix without detectable Proteins, one of the most important biomolecules in residue destruction. Cell adhesion sequences (e.g., RGD) nature, belong to this class of biopolymers. However, and enzymatic Sites have been incorporated into the poly polypeptides of a Single amino acid or copolymers of were merS for cell attachment and catalytic activity Studies, generally regarded as impractical industrial materials respectively. Synthesis utilizing genetic engineering approach has also been reported. These class of polymers (Nathan and Kohn, 1994). Amino acid N-carboxyanhydrides have been reported to exhibit excellent biocompatibility were prepared as the monomeric Starting materials, and this (Urry et al., 1995; Urry, 1993). added considerably to the cost of all polypeptides. These 15 polymers were thus expensive even if they were derived One specific polypeptide (GVGVP), has been shown to from cheap amino acids. In addition, it was almost impos undergo an inverse temperature transition in water (Urry, Sible to control the Sequence of the protein polymers using 1988a; Urry, 1988b). The mechanism of such elasticity has random copolymerization techniques. Most polypeptides are been demonstrated to be entropic in nature and is apparently insoluble in common organic Solvents. The need for exotic due to the internal chain dynamics of the ordered polypep Solvent Systems to process these materials combined with tide structure. This is contrary to the common belief that the their thermal instability made them poor engineering mate elasticity of elastin, Similar to Synthetic polymers, is due to rials. random chain network and random end-to-end distances A number of recent approaches may, however, bypass (Alberts et al., 1983). The transition temperature can be these difficulties. Advances in genetic engineering have 25 controlled by the amino acid composition, pH and enabled investigators to obtain protein polymers by inserting phosphorylation, electrochemical, photochemical and DNA templates of predetermined Sequences into the genome chemical reactions of prosthetic groups. Therefore, a device of bacteria. Collagen-like, -like, and Silk-elastin-like that converts chemical energy into mechanical work can be proteins have been Synthesized by this technique (Goldberg constructed. d. Blends, Interpenetrating Networks (IPN) and Com et al., 1989; Cappello et al., 1990; McGrath et al., 1992). posites The general concept (O'Brien, 1993) involves the incor Although there are large number of polymer blends poration of amino acid Sequences with desired properties, described in the literature, only those blends that contain e.g., cell adhesion or elasticity, into the protein polymers to biodegradable polymers and/or natural components will be produce materials of predetermined Structure and controlled 35 applicable in the context of oral tissue engineering where the properties. For example, a cell adhering Sequence of RGD matrix itself is administered along with the tissue. The use has been incorporated into Silk-like protein polymers in a of polymer blends or composites (polymeric composite manner Such that the tripeptide Sequence is exposed for cell materials) as biomaterials is a concept that nature exploits in attachment (Tirrell et al., 1994). assembling ECM in tissue. The ECM of tissues typically Investigators have also developed chemical Synthetic 40 contains a composite of different macromolecules and non techniques that are complementary to the genetic approach macro molecular material S. For example, to prepare Such materials. For instance, different rigid non glycoaminoglycans, which are usually covalently linked to peptide, organic Segments have been combined with proteins to form proteoglycans, constitutes a gel-like, highly leucine-glutamine-proline, a Sequence of the calcium bind hydrated Structure Substance in the which the collagen fibers ing domain of bovine amelogenins, using a completely 45 are embedded (Wight et al., 1991; Giusti et al., 1993). Synthetic approach (Sogah et al., 1994). The advantage of Blends of fibrin and polyurethane have previously been this class of protein-based hybrid polymers is the virtually formed by a combined phase-inversion and Spray process to unlimited choice of building blocks for the polymers. In produce highly porous Small-diameter Vascular prostheses. contrast, genetically engineered proteins can only make use (Giusti et al., 1985, Soldani et al., 1992) These materials of the 20 natural and a limited number of unnatural amino 50 exhibit high thermal stability, and their tensile behavior acids for the construction of polymers. ranged from that of an elastic polyurethane tube to that of a Rigid organic Segments have been used to reduce the natural blood vessel. Hydrogels of fibrin and poly(vinyl conformational flexibility of the peptide chain through the alcohol), blends or IPNs of collagen and poly(vinyl alcohol), formation of peptide Secondary structures (e.g., f-sheet or blends of hyaluronic acid with poly(vinyl alcohol) or poly f3-turns). Controlled folding of the polymer backbone has 55 (acrylic acid), and blends based on esters of hyaluronic acid been reported using Such ordered building blocks (Wong, have been reported (Giusti et al., 1993). These materials may 1996). The potential application of these materials to tissue be Suitable for a variety of applications including Soft tissue engineering is significant. These Synthetic techniques allow replacement, drug delivery, nerve-guide growth and cardio precise control of material properties, while maintaining the vascular devices. This class of materials has great potential freedom and flexibility to design protein-like materials with 60 owing to the large number of readily available Synthetic desirable biological and chemical properties. These proper polymers that can be mixed with biopolymers. ties may make this class of materials desirable matrices for B. Immobilization Materials tissue engineering applications. Immobilization materials are utilized to physically con Urry and coworkers have also studied elastin protein fine active biologic components (e.g., enzymes, proteins, based polymers as biocompatible materials. These 65 cell organelles and cells) while they are carrying out their polymers, also known as bioelastic materials, are elasto biological functions. The choice of matrices for cell immo meric polypeptides comprised of the general repeating bilization have been reviewed by Scouten (1995). 5,885,829 71 72 1. Polysaccharides alginate in aqueous solution (Sutherland, 1991). However, Polysaccharides are carbohydrates characterized by the exposure of alginate to Soluble calcium leads to a preferen presence of a repeating Structure in which the interunit tial binding of calcium and Subsequent gelling. These gentle linkages are of the O-glycoside type. The hydrophilicity of gelling conditions are in contrast to the large temperature or polysaccharides, along with the ease in which they can be Solvent changes typically required to induce Similar phase formed into hydrogels, makes these materials ideal for many changes in most materials. tissue engineering applications in which one desires to Alginates have been utilized as immobilization matrices immobilize cells within a matrix. The variety of Saccharides for cell (Smidsrod and Skjak-Braek, 1990), as an injectable monomers (~200) and the variety of possible O-glycoside matrix for engineering cartilaginous tissue to treat vesi linkages result in a diversity of polysaccharide Structures coureteral reflux in various animal models (Atala et al., 1993 and conformations. Polysaccharides may be derived from and Atala et al., 1994), and as injectable microcapsules different Sources including plants (starch, cellulose), animal containing islet cells to treat animal models of diabetes (Sun (glycogen), algae and Seaweeds (alginate and agarose) and et al., 1984). microorganisms. These materials are usually considered as 15 The open lattice structure and wide distribution of pore naturally-derived products. However, Since polysaccharides sizes in calcium alginate preclude the controlled release of are widely utilized as immobilization materials, they are large molecules (e.g., proteins) from these materials and used here as Standards to which other Synthetic materials are limits the use of pure alginate for entrapment of whole cells compared. or cell organelles (Smidsrod and Skjak-Braek, 1990). a. Algal Polysaccharides: Alginate and Agarose However, alginate membrane can be modified by incorpo Algal polysaccharides have been the most commonly rating other polymeric elements (e.g., lysine, poly(ethylene utilized immobilization materials. This is due to their gentle glycol), poly(vinyl alcohol) or chitosan) (Polk et al., 1994, gelling conditions, widespread availability, and relative bio Kung et al., 1995). These modified systems have been used compatibility. The main Starting Sources of alginate are 25 to control the release of proteins from alginate beads. Species of brown algae (Phaeophyceae). The algae are Haemostatic Swabs made of calcium alginate have also been typically Subjected to a number of processing Steps to clinically utilized to reduce blood loSS during Surgical pro produce pure alginate which is the major polysaccharide cedures. The calcium ions in alginate may assist the blood present and may comprise up to 40% of the dry weight. It is clotting process by activating platelets and clotting factor part of the intracellular matrix and exists, in the native State, as a mixed Salt of the various cations found in Sea water VII (Blair et al., 1990). (e.g., Mg, Ca, Sr., Ba and Na"). Due to selectivity of Agarose is another type of marine algal polysaccharide. In cation binding, the native alginate is mainly found in the contrast to alginate, agarose forms thermally reversible gels. insoluble gel form, which results from cross-linking of Agarose will Set at concentrations in excess of 0.1%, alginate chains by Ca". 35 depending on the Sulfate content, and at temperatures con All alginates are copolymers of D-mannuronate (M) and siderably below (~40°C.) the gel-melting temperature (~90 L-guluronate (G). However, alginates from different algal C.). The latter parameter is correlated to the methoxy Sources have different compositions, and thus, different content. The proposed gel Structure is bundles of associated physical and mechanical properties. The block length of 40 double helices and the junction Zones consist of multiple monomer units, and overall composition of the alginate and chain aggregations (Yalpani, 1988). Agarose has been used molecular weight, determine the properties of alginates. For largely in gels for electrophoresis of proteins and nucleic example, calcium alginates rich in G are Stiff materials acids. However, agarose gels have also been used as Sup (Sutherland, 1991). porting materials for electrophoresis of bacteriophages Alginate Selectively binds divalent metal ions Such as 45 (Serwer, 1987) and migration studies of leukocytes (Kallen Ba", Sr" and Ca". The binding selectivity increases with et al., 1977). Although applications in tissue engineering G content, and polymannuronate is essentially non have not been reported, its adjustable gelling behavior may Selective. The calcium ions are, therefore, Selectively bound render low temperature melting agarose a Suitable injectable between Sequences of polyguluronate residue, and are held and immobilization matrix material. between diaxially linked L-guluronate residues which are in 50 b. Other Polysaccharides the C chair conformation. The calcium ions are thus Microbial polysaccharides are ubiquitous in nature and packed into the interstices between polyguluronate chains very abundant biopolymers. They are of interest because of asSociated pairwise and this structure is named the "egg box” sequence. The ability to form a junction Zone depends their unusual and useful functional properties. Some of these on the length of the G-blocks in different alginates. Since the 55 properties are Summarized by Kaplan et al. (1994) as: (i) mechanical Strength of alginate gels depend on the block film-forming and gel-forming capabilities, (ii) Stability over lengths and M/G content, there have been efforts to modify broad temperature ranges, (iii) biocompatibility (natural the M/G ratio by alginase to increase the G content (Skjak products avoid the release/leaching of toxic metals, residual chemicals, catalyst, or additives), (iv) unusual rheological Braek et al., 1986). It is expected that chemically modified 60 alginate would also produce materials of desirable proper properties, (v) biodegradability, (vi) water Solubility in the ties. For example, bacterial alginates that contains acetyl native state or reduced solubility if chemically modified, and groups generally exhibit different physical and mechanical (vii) thermal processability for Some of these polymers. properties from those of algal sources (Ott and Day, 1995). Some examples of microbial polysaccharide are listed in Alginate can be gelled under mild conditions, allowing 65 Table 9. It is worthy to note that gellan, one of the microbial cell immobilization with little damage. Binding of Mg" and polysaccharides, has been investigated as immobilization monovalent ions to alginate does not induce gelation of materials for enzymes and cells (Doner and Douds, 1995). 5,885,829 73 74 Lawson, 1960). However, a variety of complications were TABLE 9 found after long term implantation, including calcification of the PVA (Peters and Smith, 1981). Some Polysaccharides Synthesized by Microorganisms More recently, PVA was made into an insoluble gel using Polymers' Structure a physical cross-linking process. These gels were prepared with a repeated freezing-thawing proceSS. This causes Struc Fungal tural densification of the hydrogel due to the formation of Pullulan (N) 1,4-1,6-O-D-Glucan Semicrystalline Structures. The use of this gel in drug deliv Scleroglucan (N) 1,3-1,6-O-D-Glucan ery applications has been reported (Peppas and Scott, 1992; Chitin (N) 1,4-f-D-Acetylglucosamine Ficek and Peppas, 1993). However, PVA is not truly biode Chitosan (C) 1,4-B-D-N-Glucosamine gradable due to the lack of labile bonds within the polymer Elsinan (N) 1,4-1,3-C-D-Glucan bond. Only low molecular weight materials are advisable to Bacterial be used as implant materials. Xanthan gum (A) 1,4-f-D-Glucan with D-mannose: D-glucuronic acid as c. Poly(ethylene oxide) (PEO) side groups 15 PEO or polyethylene glycol can be produced by the Curdlan (N) 1,3-f-D-Glucan (with branching) Dextran (N) 1,6-O-D-Glucan with some 1.2-1,3-1,4-O-linkages anionic or cationic polymerization of ethylene oxide using a Gellan (A) 1,4-f-D-Glucan with rhamose, D-glucuronic acid variety of initiators (Boileau, 1989; Penczek and Kubisa, Levan (N) 2,6-f-D-Fructan with some ?-2,1-branching 1989). PEO is highly hydrophilic and biocompatible, and Emulsan (A) Lipoheteropolysaccharide has been utilized in a variety of biomedical applications Cellulose (N) 1,4-B-D-Glucan including preparation of biologically relevant conjugates N = neutral, A = anionic and C = cationic. (Zalipsky, 1995), induction of cell membrane fusion (Lentz, Adapted from Wong and Mooney, 1997. 1994) and surface modification of biomaterials (Amiji and 2. Non-Natural Hydrogels Park, 1993). Different polymer architectures have been a. PolyphosphaZenes Synthesized and Some of their applications in medicine have PolyphosphaZenes contain inorganic backbones com 25 been recently reviewed (Merrill, 1993). For example, PEO prised of alternating Single and double bonds between can be made into hydrogels by Y-ray or electron beam nitrogen and phosphorus atoms, in contrast to the carbon irradiation and chemical crosslinking (Cima et al., 1995; carbon backbone in most other polymers. The uniqueness of Belcheva et al., 1996). These hydrogels have been used as polyphosphaZenes Stems from the combination of this inor matrices for drug delivery and cell adhesion Studies. ganic backbone with Versatile side chain functionalities that d. Pluronics can be tailored for different applications. The degradation of Pluronic polyols or polyoxamers are block copolymers of polyphosphaZenes results in the release of phosphate and PEO and poly(propylene oxide and are usually Synthesized ammonium ions along with the side groups (Allcock, 1989; by anionic polymerization in the form of a ABA triblock Scopelianos, 1994). using a difunctional initiator (Schmolka, 1972). Pluronics F Linear, uncroSS-linked polymers can be prepared by ther 35 127, which contains 70% ethylene oxide and 30% propylene mal ring opening polymerization of (NPCl) and the chloro oxide by weight with an average molecular weight of group replaced by amines, alkoxides or organometallic 11,500, is the most commonly used gel-forming polymer reagents to form hydrolytically Stable, high molecular matrix to deliver proteins (Gombotz and Pettit, 1995). weight poly(organophosphaZenes). Depending on the prop This polymer exhibits a reversible thermal gelation in erties of the Side groups, the polyphosphaZenes can be 40 aqueous Solutions at a concentration of 20% or more hydrophobic, hydrophilic or amphiphilic. The polymers can (Schmolka, 1972). Thus, the polymer solution is a liquid at be fabricated into films, membranes and hydrogels for room temperature but gels rapidly in the body. Although the biomedical applications by cross-linking or grafting (Lora et polymer is not degraded by the body, the gels dissolve al., 1991; Allcock et al., 1988; Allcock, 1989). Bioerodible Slowly and the polymer is eventually cleared. This polymer polymers for drug delivery devices have been prepared by 45 has been utilized in protein delivery (Morikawa et al., 1987; incorporating hydrolytic Side chains of imidazole Jushasz et al., 1989) and skin burn treatments (Pautian et al., (Laurencin et al., 1987) for skeletal tissue regeneration 1993). (Laurencinet al., 1993). Non-degradable phosphazenes have e. PGA-PEO Hydrogels been used as denture liner (Razavi et al., 1993). Their use in Although PGA is not water soluble, bioerodible hydrogels the present invention is thus particularly contemplated. 50 based on photopolymerized PGA-PEO copolymers have b. Poly(vinyl alcohol) (PVA) been Synthesized and their biological activities investigated. PVA is not synthesized directly but is the deacetylated (Sawhney et al., 1993; Sawhney et al., 1994; Hill-West et al., product of poly(Vinyl acetate). Polyvinyl acetate is usually 1994). Macromonomers having a poly(ethylene glycol) cen prepared by radical polymerization of vinyl acetate (bulk, tral block, extended with oligomers of C-hydroxy acids (e.g., solution or emulsion polymerizations) (Finch, 1973). PVA is 55 oligo(dl-lactic acid) or oligo(glycolic acid)) and terminated formed by either alcoholysis, hydrolysis or aminolysis pro with acrylate groups were Synthesized. These hydrogels cesses of poly(Vinyl acetate). The hydrophilicity and water were designed to form direct contacts with tissueS or pro solubility of PVA can be readily controlled by the extent of teins following photopolymerization, and act as a barrier. hydrolysis and molecular weight. PVA has been widely used These gels degrade upon hydrolysis of the oligo(C- as thickening and Wetting agent. 60 hydroxy acid) regions into poly(ethylene glycol), the PVA gels can be prepared by croSS-linking with formal O-hydroxy acid, and oligo(acrylic acid). The degradation dehyde in the presence of Sulfuric acid (Schwartz et al., rate of these gels could be tailored from leSS than 1 day to 1960). These formaldehyde-cross-linked PVA materials 4 months by appropriate choice of the oligo(C.-hydroxy have been used as prosthesis for a variety of plastic Surgery acid). The macromonomer could be polymerized using applications including breast augmentation (Clarkson, 1960 65 non-toxic photoinitiators with visible light without exceSS and Peters and Smith, 1981), diaphragm replacement (Haupt heating or local toxicity. The hydrogels polymerized in and Myers, 1960) and bone replacement (Camerson and contact with tissue adhere tightly to the underlying tissue. In 5,885,829 75 76 contrast, the gels were nonadhesive if they were polymer Schmitt, 1971), and are generally considered to be biocom ized prior to contact with tissue. These hydrogels have been patible. The crystallinity, mechanical properties, and erosion utilized in animal models to prevent post-Surgical adhesion times of these polymerS is regulated by the ratio of lactic:g- and thrombosis of blood vessels and initimal thickening lycolic acid (Gilding, 1981). This example describes the following balloon catheterization. 5 fabrication and characterization of tubular cell delivery It can thus be seen that there are a large number of devices from a range of polymers of the lactic and glycolic Synthetic biodegradable polymers that may be used in the acid. Devices with a wide range of compression resistance oral tissue engineering invention described herein. Estab and erosion times can be fabricated by utilizing PLA, lished polymer chemistries enable one to tailor properties of PDLLA, and PLGA. the Synthetic polymers by using different i) functional A. Materials groups (either on the backbone or side chain), ii) polymer PLA, PDLLA, and the 85/15 and 50/50 PLGA were architectures (linear, branched, comb or star), and iii) com purchased from Medisorb (Cincinnati, Ohio), chloroform binations of polymer species physically mixed (polymer from Mallinckrodt (Paris, Ky.), polystyrene standards from blends or interpenetrating networks) or chemically bonded PolySciences (Warrington, Pa.), aluminum backed tape from (copolymers) (Gombotz and Pettit, 1995). The current pref 15 Cole-Parmer (Chicago, Ill.), phosphate buffered saline and erence for PGA and related polyesters is partially due to their DMEM medium from Gibco (Grand Island, N.Y.), Tmax established Safety in human applications, and the projected film from Kodak, Lewis rats, 250 to 300 g, from Charles approval of the Food and Drug Administration. PLGA can River (Wilmington, Mass.), and methoxyflurane from also be used with Specific peptide Sequences incorporated Pitman-Moore Inc. (Mundelein, Ill.). into the polymer. Polymers constituted of building blocks Molecular weights of the various polymers were deter Similar to components of ECM, e.g., carbohydrates and mined by gel permeation chromatography (Perkin-Elmer, peptides, may also be used. Series 10, Newton Centre, Mass.), using polystyrene stan dards to generate a calibration curve. PLA had a molecular EXAMPLE V weight (M) of 74,000 (M/M=1.6); poly-(D.L. lactic) acid Fabricating Tubular Matrices 25 had M=77,000 (M/M=1.8); 85/15 copolymer had M=69,000 (M/M=1.9); 50/50 copolymer M=43,400 AS described above, polymers of lactic and glycolic acid (M/M=1.43). Differential scanning calorimetry was ulti are attractive candidates to fabricate devices to transplant lized to confirm the amorphous nature of all of the polymers cells and engineer new tissues. These polymers are except PLLA, which exhibited the expected crystallinity. biocompatible, and exhibit a wide range of erosion times and B. Device Fabrication mechanical properties. This example describes the fabrica Hollow tubes were formed by a two-step process, porous tion and characterization, in vitro and in Vivo, of hollow, films of the polymers were first fabricated, and these films tubular devices from porous films of various polymers of were then formed into hollow tubes. To fabricate porous this family. films, the polymer was dissolved in chloroform to form a Porous films of these polymers were formed using a 35 1.56% solution (w/v). Eight ml of this solution was cast into particulate leaching technique, and Sealed around teflon a 5 cm glass petri dish covered with a sheet of aluminum cylinders to form hollow tubular devices. The erosion rate of backed tape. Sieved sodium chloride crystals (150

UNITED STATES PATENT AND TRADEMARK OFFICE CERTIFICATE OF CORRECTION

PATENT NO. 5,885,829 DATED : March 23, 1999 INVENTOR(S) : Mooney et al. it is certified that error appears in the above-identified patent and that said Letters Patent is hereby Corrected as shown below: On the title page, item 51, line 1 through 2, delete Cl2N 5700; C12N 5 02: Cl2N 5 08: Ci 2N 15 09' and insert -- C2N 5/06, 5/08, 5/10, A6 L 27/00, A61K 6700 --

Signed and Sealed this Twenty-sixth Day of October, 1999 Aftest: 2.76%

Q. TOD) DECKNSON Attesting Officer A 'ting Commissionei (f Patents and Tracientirks