Bringing Safe and Standardized Cell Therapies to Industrialized Processing for Burns and Wounds

Alexis Laurent, Poyin Lin, Corinne Scaletta, Nathalie Hirt-Burri, Murielle Michetti, Anthony S de Buys Roessingh, Wassim Raffoul, Bin-Ru She, Lee Ann Applegate, Alexis Laurent, Poyin Lin, Corinne Scaletta, Nathalie Hirt-Burri, Murielle Michetti, Anthony S de Buys Roessingh, Wassim Raffoul, Bin-Ru She, Lee Ann Applegate

Abstract

Cultured primary progenitor cell types are worthy therapeutic candidates for regenerative medicine. Clinical translation, industrial transposition, and commercial implementation of products based on such cell sources are mainly hindered by economic or technical barriers and stringent regulatory requirements. Applied research in allogenic cellular therapies in the Lausanne University Hospital focuses on cell source selection technique optimization. Use of fetal progenitor cell sources in Switzerland is regulated through Federal Transplantation Programs and associated Fetal Biobanks. Clinical applications of cultured primary progenitor dermal fibroblasts have been optimized since the 1990s as "Progenitor Biological Bandages" for pediatric burn patients and adults presenting chronic wounds. A single organ donation procured in 2009 enabled the establishment of a standardized cell source for clinical and industrial developments to date. Non-enzymatically isolated primary dermal progenitor fibroblasts (FE002-SK2 cell type) served for the establishment of a clinical-grade Parental Cell Bank, based on a patented method. Optimized bioprocessing methodology for the FE002-SK2 cell type has demonstrated that extensive and consistent progenitor cell banks can be established. In vitro mechanistic characterization and in vivo preclinical studies have confirmed potency, preliminary safety and efficacy of therapeutic progenitor cells. Most importantly, highly successful industrial transposition and up-scaling of biobanking enabled the establishment of tiered Master and Working Cell Banks using Good Manufacturing Practices. Successive and successful transfers of technology, know-how and materials to different countries around the world have been performed. Extensive developments based on the FE002-SK2 cell source have led to clinical trials for burns and wound dressing. Said trials were approved in Japan, Taiwan, USA and are continuing in Switzerland. The Swiss Fetal Transplantation Program and pioneer clinical experience in the Lausanne Burn Center over three decades constitute concrete indicators that primary progenitor dermal fibroblasts should be considered as therapeutic flagships in the domain of wound healing and for regenerative medicine in general. Indeed, one single organ donation potentially enables millions of patients to benefit from high-quality, safe and effective regenerative therapies. This work presents a technical and translational overview of the described progenitor cell technology harnessed in Switzerland as cellular therapies for treatment of burns and wounds around the globe.

Keywords: GMP manufacturing; burns; cell therapy; chronic wounds; clinical cell banking; fibroblasts; progenitor cells.

Copyright © 2020 Laurent, Lin, Scaletta, Hirt-Burri, Michetti, de Buys Roessingh, Raffoul, She and Applegate.

Figures

Figure 1
Figure 1
Technological advantages of appropriate whole-cell bioprocessing for skin progenitors. From one single organ donation (FE002, 2009), various samples yielding differentiated tissue-specific progenitor cells were bioprocessed for isolation using the proprietary non-enzymatic method. Intrinsic cellular identity and characteristics were therein optimally maintained throughout the transposition to adherent monolayer culture. Inherent technical and clinical advantages are attributed to the specific choice of the cell source. Optimized and consistent biobanking allowed for establishment of extensive GMP cell banks. Thorough testing throughout manufacturing along with the consistency of the cell source guarantees optimal homogeneity and safety of the biological starting material for therapeutic products. Hundreds of millions of treatments can be produced based on the available stocks.
Figure 2
Figure 2
FE002-SK2 stock projections and product manufacture. Assuming maximal expansion of all available cells, the theoretical number of final products is quantified in dozens of billions. Quality and safety testing are highly consumptive in materials throughout the GMP manufacturing process, but further optimization of bioprocessing and biobanking protocols would reasonably allow for such numbers of effective treatments to be produced, using one dedicated organ donation. To manufacture end-products, cryopreserved FE002-SK2 cells at specific passages are thawed and seeded on sterile bio-compatible scaffolds, which are applied to patients following defined clinical protocols.
Figure 3
Figure 3
FE002-SK2 GMP biobank establishment and validation. After the FE002-SK2 PCB was declared fit for GMP manufacturing, expansion campaigns were conducted for the establishment of extensive and consistent GMP FE002-SK2 Master (P2), Working (P4), and End of Production (P12) cell banks. All the progeny cells were stored in liquid nitrogen vapor phase. Several vials of each production batch were initiated for recovery studies and quality release testing. The EOPCB materials served for extensive safety and stability assays, both in vitro and in vivo. The complete battery of tests comprised (a) isoenzyme tests, (b) sterility tests, (c) mycoplasma detection, (d) karyology, (e) transmission electron microscopy (viruses, virus-like particles, mycoplasma, yeasts, fungi, bacteria), (f) in vitro adventitious viruses testing (picornavirus, orthomyxovirus, pramyxovirus, herpesvirus, adenovirus, reovirus, West Nile virus), (g) in vivo adventitious viruses testing (suckling mice, adult mice, guinea pigs, embryonated eggs), (h) Q-PCR (HepB/C, HIV-1/2, HTLV-1/2, HHV-6/7/8, EBV, hCMV, SV40, B19 parovirus) and (i) in vivo tumorigenicity testing in nude mice.
Figure 4
Figure 4
FE002-SK2 tiered biobanking and GMP tec-transfers. The robust biobanking protocols devised for the FE002-SK2 progenitor fibroblasts allowed for successful technology transfers to BioReliance (Scotland), Transwell Biotech Co. Ltd. (Taiwan), and the CHUV CPC (Epalinges/Lausanne, Switzerland). Varying banking nomenclatures exist between the different groups, the pertinent passage numbers being those used by BioReliance. Quantities of vials listed exclude those consumed for recovery studies and release testing. Pre-clinical safety testing was performed on the EOPCB by BioReliance. Clinical applications comprise the ongoing use of PBBs in the CHUV Burn Center, the pending Priority Project Bru_PBB and both clinical trials sponsored in Asia by TWB.
Figure 5
Figure 5
Representative imaging of migrated primary human keratinocytes (DAPI staining) in Transwell® migration assay (left and middle columns). Representative imaging includes middle fields of 3 wells/group. Contamination of Transwells® by FE002-SK2 progenitor fibroblasts was negligible (right column). Mean cell counts are listed in Table 1.
Figure 6
Figure 6
Epithelial migration in the presence of FE002-SK2 conditioned media and sham media. Quantitative results of three experimental repetitions are compared (A–C). At the 9- and 18-h timepoints in the second and third replicates, respectively (B,C), the gaps treated with FE002-SK2 conditioned media were completely filled. Representative imaging of epithelial migration from the first experiment is shown (D). Negligible filling was detectable at the 3-h timepoint. Imaging data was acquired under 40X optical magnification.
Figure 7
Figure 7
Representative imaging of keratinocyte proliferation in the absence and presence of FE002-SK2 cell extracts. Adjunction of sham medium or cell extracts on Day 0 did not stimulate keratinocyte proliferation and did not ensure maintenance of cell populations (A–F,J–L). Adjunction of cell extracts on Day 0 and Day 2 resulted in augmented keratinocyte proliferation (H,I,N,O). Imaging data was acquired under 40X optical magnification. Mean harvesting total cell counts are listed in Table 2.
Figure 8
Figure 8
Proliferation of primary keratinocytes in the presence of mitomycin C-treated FE002-SK2 progenitor fibroblasts (MMC-FE002-SK2 cells). DK-SFM is deficient in extracellular matrix (ECM) proteins. Considering that FE002-SK2 fibroblasts can secrete ECM proteins for cell attachment, both collagen-coated (A–I) and un-coated surfaces (J–S) were comparatively tested. No cell proliferation was observed in the single-population cultures of keratinocytes (D–F,J–L) or MMC-FE002-SK2 cells (A–C). Keratinocyte proliferation was observable in co-cultures (G–I,M–P). Selected wells were also stained with anti-CK-14 antibodies (P–S). MMC-FE002-SK2 progenitor fibroblasts appeared negative for immunostaining (S), while proliferating cells in co-culture wells appeared positive (Q). Staining data confirmed that proliferating cells were keratinocytes. Imaging data was acquired under 40X optical magnification and fluorescence microscopy.
Figure 9
Figure 9
Representative imaging of primary keratinocyte and FE002-SK2 progenitor fibroblast proliferation in single- and co-cultures. Initial viable seeding densities are indicated for each FE002-SK2 group. Initial viable seeding density of keratinocyte was of 2,000 cells/cm2. No proliferation was observed in the single-population cultures of keratinocytes (A–D) or FE002-SK2 fibroblasts (E–L). High proliferation was observable in the co-cultures (M–T). Imaging data was acquired under 40X optical magnification. Mean harvesting cumulated cell counts are listed in Table 3.
Figure 10
Figure 10
Representative and evolutive imaging of porcine wound models. Data were acquired from pig N°052. All 4 experimental groups are presented at different timepoints ranging from wound creation (Day 0) up until sacrifice (Day 14). Treatment groups [A–D] consisted in the sham control, hydrogel scaffold alone, and hydrogel scaffolds with low and high doses of FE002-SK2 progenitor fibroblasts, respectively.
Figure 11
Figure 11
Representative imaging of tissue sections isolated for histological examination. Sections were isolated on Day 14 and stained using HE. Treatment groups [A–D] consisted in the sham control (A), hydrogel scaffold alone (B) and hydrogel scaffolds with low (C) and high doses (D) of FE002-SK2 progenitor fibroblasts, respectively. Arrows in (A,B) indicate observable epidermal detachment.

References

    1. Abbasalizadeh S., Baharvand H. (2013). Technological progress and challenges towards cGMP manufacturing of human pluripotent stem cells based therapeutic products for allogeneic and autologous cell therapies. Biotechnol. Adv. 31, 1600–1623. 10.1016/j.biotechadv.2013.08.009
    1. Abbasalizadeh S., Pakzad M., Cabral J. M. S., Baharvand H. (2017). Allogeneic cell therapy manufacturing: process development technologies and facility design options. Expert Opin. Biol. Ther. 17, 1201–1219. 10.1080/14712598.2017.1354982
    1. Abdel-Sayed P., Hirt-Burri N., de Buys Roessingh A. S., Raffoul W., Applegate L. A. (2019). Evolution of biological bandages as first cover for burn patients. Adv. Wound Care 8, 555–564. 10.1089/wound.2019.1037
    1. Abdel-Sayed P., Kaeppeli A., Siriwardena T., Darbre T., Perron K., Jafari P., et al. (2016). Anti-microbial dendrimers against multidrug-resistant P. aeruginosa enhance the angiogenic effect of biological burn-wound bandages. Sci. Rep. 6:22020 10.1038/srep23872
    1. Akita S., Akino K., Imaizumi T., Hirano A. (2008). Basic fibroblast growth factor accelerates and improves second-degree burn wound healing. Wound Repair Regen. 16, 635–641. 10.1111/j.1524-475X.2008.00414.x
    1. Antsiferova M., Klatte J. E., Bodó E., Paus R., Jorcano J. L., Matzuk M. M., et al. . (2009). Keratinocyte-derived follistatin regulates epidermal homeostasis and wound repair. Lab. Invest. 89, 131–141. 10.1038/labinvest.2008.120
    1. Applegate L. A., Scaletta C., Hirt-Burri N., Raffoul W., Pioletti D. (2009). Whole-cell bioprocessing of human fetal cells for tissue engineering of skin. Skin Pharmacol. Physiol. 22, 63–73. 10.1159/000178865
    1. Applegate L. A., Weber D., Simon J. P., Scaletta C., Hirt-Burri N., de Buys Roessingh A. S., et al. . (2013). Organ donation and whole-cell bioprocessing in the Swiss fetal progenitor cell transplantation platform, in Organ Donation and Organ Donors. ed Saidi R. F. (Nova Science Publishers; ), 125–147.
    1. Bach A. D., Arkudas A., Tjiawi J., Polykandriotis E., Kneser U., Horch R. E., et al. . (2006). A new approach to tissue engineering of vascularized skeletal muscle. J. Cell. Mol. Med. 10, 716–726. 10.1111/j.1582-4934.2006.tb00431.x
    1. Barrientos S., Stojadinovic O., Golinko M. S., Brem H., Tomic-Canic M. (2008). Growth factors and cytokines in wound healing. Wound Repair Regen. 16, 585–601. 10.1111/j.1524-475X.2008.00410.x
    1. Borda L. J., Macquhae F. E., Kirsner R. S. (2016). Wound dressings: a comprehensive review. Curr. Derm. Rep. 5, 287–297. 10.1007/s13671-016-0162-5
    1. Cass D. L., Meuli M., Adzick N. S. (1997). Scar wars: implications of fetal wound healing for the pediatric burn patient. Pediatr. Surg. Int. 12, 484–489. 10.1007/BF01258707
    1. Chen H. L., Chiang P. C., Lo C. H., Lo Y. H., Hsu D. K., Chen H. Y., et al. . (2016). Galectin-7 regulates keratinocyte proliferation and differentiation through JNK-miR-203-p63 signaling. J. Invest. Dermatol. 136, 182–191. 10.1038/JID.2015.366
    1. Costa-Almeida R., Calejo I., Gomes M. E. (2019). Mesenchymal stem cells empowering tendon regenerative therapies. Int. J. Mol. Sci. 20:3002. 10.3390/ijms20123002
    1. De Buys Roessingh A. S., Hirt-Burri N., Raffoul W., Scaletta C., Applegate L. A. (2015). A decade after foetal skin progenitor cell therapy in pediatric burn treatment. J. Regen. Med. 4:1 10.4172/2325-9620.1000122
    1. De Buys Roessingh A. S., Hohlfeld J., Scaletta C., Hirt-Burri N., Gerber S., Hohlfeld P., et al. . (2006). Development, characterization, and use of a fetal skin cell bank for tissue engineering in wound healing. Cell Transplant. 15, 823–834. 10.3727/000000006783981459
    1. Dhivya S., Padma V. V., Santhini E. (2015). Wound dressings - a review. Biomedicine 5:22. 10.7603/s40681-015-0022-9
    1. Doyle A., Griffiths J. B. (1998). Cell and Tissue Culture: Laboratory Procedures in Biotechnology. New York, NY: Wiley & Sons.
    1. Gallucci R. M., Simeonova P. P., Matheson J. M., Kommineni C., Guriel J. L., Sugawara T., et al. . (2000). Impaired cutaneous wound healing in interleukin-6-deficient and immunosuppressed mice. FASEB J. 14, 2525–2531. 10.1096/fj.00-0073com
    1. Grognuz A., Scaletta C., Farron A., Raffoul W., Applegate L. A. (2016). Human fetal progenitor tenocytes for regenerative medicine. Cell Transplant. 25, 463–479. 10.3727/096368915X688515
    1. Haack-Sørensen M., Kastrup J. (2011). Cryopreservation and revival of mesenchymal stromal cells. Methods Mol. Biol. 698, 161–174. 10.1007/978-1-60761-999-4_13
    1. Hayflick L., Plotkin S. A., Norton T. W., Koprowski H. (1962). Preparation of poliovirus vaccines in a human fetal diploid cell strain. Am. J. Hyg. 75, 240–258. 10.1093/oxfordjournals.aje.a120247
    1. Heathman T. R., Nienow A. W., McCall M. J., Coopman K., Kara B., Hewitt C. J. (2015). The translation of cell-based therapies: clinical landscape and manufacturing challenges. Regen. Med. 10, 49–64. 10.2217/rme.14.73
    1. Hebda P. A., Dohar J. E. (1999). Transplanted fetal fibroblasts: Survival and distribution over time in normal adult dermis compared with autogenic, allogenic, and xenogenic adult fibroblasts. Otolaryngol. Head Neck Surg. 121, 245–251. 10.1016/S0194-5998(99)70179-8
    1. Hernon C. A., Dawson R. A., Freedlander E., Short R., Haddow D. B., Brotherston, et al. (2006). Clinical experience using cultured epithelial autografts leads to an alternative methodology for transferring skin cells from the laboratory to the patient. Regen. Med. 1, 809–821. 10.2217/17460751.1.6.809
    1. Hohlfeld J., de Buys Roessingh A. S., Hirt-Burri N., Chaubert P., Gerber S., Scaletta C., et al. . (2005). Tissue engineered fetal skin constructs for paediatric burns. Lancet 366, 840–842. 10.1016/S0140-6736(05)67107-3
    1. Hunsberger J., Harrysson O., Shirwaiker R., Starly B., Wysk R., Cohen P., et al. . (2015). Manufacturing road map for tissue engineering and regenerative medicine technologies. Stem Cells Transl. Med. 4, 130–135. 10.5966/sctm.2014-0254
    1. Hunt C. J. (2019). Technical considerations in the freezing, low-temperature storage and thawing of stem cells for cellular therapies. Transfus. Med. Hemother. 46, 134–150. 10.1159/000497289
    1. Jiang W. G., Sanders A. J., Ruge F., Harding K. G. (2012). Influence of interleukin-8 (IL-8) and IL-8 receptors on the migration of human keratinocytes, the role of PLC-γ and potential clinical implications. Exp. Ther. Med. 3, 231–236. 10.3892/etm.2011.402
    1. Kim H. R., Kim J., Park S. R., Min B. H., Choi B. H. (2018). Characterization of human fetal cartilage progenitor cells during long-term expansion in a xeno-free medium. Tissue Eng. Regen. Med. 15, 649–659. 10.1007/s13770-018-0132-z
    1. Krishnaswamy V. R., Mintz D., Sagi I. (2017). Matrix metalloproteinases: The sculptors of chronic cutaneous wounds. Biochim. Biophys. Acta Mol. Cell Res. 1864(Pt. B), 2220–2227. 10.1016/j.bbamcr.2017.08.003
    1. Kroeze K. L., Boink M. A., Sampat-Sardjoepersad S. C., Waaijman T., Scheper R. J., Gibbs S. (2012). Autocrine regulation of re-epithelialization after wounding by chemokine receptors CCR1, CCR10, CXCR1, CXCR2, and CXCR3. J. Invest. Dermatol. 132, 216–225. 10.1038/jid.2011.245
    1. Larijani B., Ghahari A., Warnock G. L., Aghayan H. R., Goodarzi P., Falahzadeh K., et al. . (2015). Human fetal skin fibroblasts: extremely potent and allogenic candidates for treatment of diabetic wounds. Med. Hypotheses 84, 577–579. 10.1016/j.mehy.2015.03.004
    1. Laurent-Applegate L. A. (2012). Preparation of Parental Cell Bank from Foetal Tissue. European Patent No 2,732,030,B1. Munich: European Patent Office.
    1. Li Z., Maitz P. (2018). Cell therapy for severe burn wound healing. Burns Trauma 6:13. 10.1186/s41038-018-0117-0
    1. Loebel C., Burdick J. A. (2018). Engineering stem and stromal cell therapies for musculoskeletal tissue repair. Cell Stem Cell. 22, 325–339. 10.1016/j.stem.2018.01.014
    1. Marks P., Gottlieb S. (2018). Balancing safety and innovation for cell-based regenerative medicine. N. Engl. J. Med. 378, 954–959. 10.1056/NEJMsr1715626
    1. Metcalfe A. D., Ferguson M. W. (2008). Skin stem and progenitor cells: using regeneration as a tissue-engineering strategy. Cell. Mol. Life Sci. 65, 24–32. 10.1007/s00018-007-7427-x
    1. Mirmalek-Sani S. H., Tare R. S., Morgan S. M., Roach H. I., Wilson D. I., Hanley N. A., et al. . (2006). Characterization and multipotentiality of human fetal femur-derived cells: implications for skeletal tissue regeneration. Stem Cells 24, 1042–1053. 10.1634/stemcells.2005-0368
    1. Monti M., Perotti C., Del Fante C., Cervio M., Redi C. A. (2012). Stem cells: sources and therapies. Biol. Res. 45, 207–214. 10.4067/S0716-97602012000300002
    1. Montjovent M. O., Burri N., Mark S., Federici E., Scaletta C., Zambelli P. Y., et al. . (2004). Fetal bone cells for tissue engineering. Bone 35, 1323–1333. 10.1016/j.bone.2004.07.001
    1. Mount N. M., Ward S. J., Kefalas P., Hyllner J. (2015). Cell-based therapy technology classifications and translational challenges. Philos. Trans. R. Soc. Lond. B Biol. Sci. 370:20150017. 10.1098/rstb.2015.0017
    1. Olshansky S. J., Hayflick L. (2017). The role of the WI-38 cell strain in saving lives and reducing morbidity. AIMS Public Health 4, 127–138. 10.3934/publichealth.2017.2.127
    1. Pastar I., Stojadinovic O., Yin N. C., Ramirez H., Nusbaum A. G., Sawaya A., et al. . (2014). Epithelialization in wound healing: a comprehensive review. Adv. Wound Care 3, 445–464. 10.1089/wound.2013.0473
    1. Pigeau G. M., Csaszar E., Dulgar-Tulloch A. (2018). Commercial scale manufacturing of allogeneic cell therapy. Front. Med. 5:233. 10.3389/fmed.2018.00233
    1. Providence K. M., Higgins S. P., Mullen A., Battista A., Samarakoon R., Higgins C. E., et al. . (2008). SERPINE1 (PAI-1) is deposited into keratinocyte migration “trails” and required for optimal monolayer wound repair. Arch. Dermatol. Res. 300, 303–310. 10.1007/s00403-008-0845-2
    1. Quintin A., Hirt-Burri N., Scaletta C., Schizas C., Pioletti D. P., Applegate L. A. (2007). Consistency and safety of cell banks for research and clinical use: preliminary analysis of fetal skin banks. Cell Transplant. 16, 675–684. 10.3727/000000007783465127
    1. Ramelet A. A., Hirt-Burri N., Raffoul W., Scaletta C., Pioletti D. P., Offord E., et al. . (2009). Chronic wound healing by fetal cell therapy may be explained by differential gene profiling observed in fetal versus old skin cells. Exp. Gerontol. 44, 208–218. 10.1016/j.exger.2008.11.004
    1. Ratcliffe E., Thomas R. J., Williams D. J. (2011). Current understanding and challenges in bioprocessing of stem cell-based therapies for regenerative medicine. Br. Med. Bull. 100, 137–155. 10.1093/bmb/ldr037
    1. Rayment E. A., Williams D. J. (2010). Concise review: mind the gap: challenges in characterizing and quantifying cell- and tissue-based therapies for clinical translation. Stem Cells 28, 996–1004. 10.1002/stem.416
    1. Rheinwald J. G., Green H. (1975). Serial cultivation of strains of human epidermal keratinocytes: the formation of keratinizing colonies from single cells. Cell 6, 331–343. 10.1016/S0092-8674(75)80001-8
    1. Scholz F., Schulte A., Adamski F., Hundhausen C., Mittag J., Schwarz A., et al. . (2007). Constitutive expression and regulated release of the transmembrane chemokine CXCL16 in human and murine skin. J. Invest. Dermatol. 127, 1444–1455. 10.1038/sj.jid.5700751
    1. Shah M., Foreman D. M., Ferguson M. W. J. (1992). Control of scarring in adult wounds by neutralising antibody to transforming growth factor β. Lancet 339, 213–214. 10.1016/0140-6736(92)90009-R
    1. Simman R., Phavixay L. (2011). Split-thickness skin grafts remain the gold standard for the closure of large acute and chronic wounds. J. Am. Col. Certif. Wound Spec. 3, 55–59. 10.1016/j.jcws.2012.03.001
    1. Sobel K., Tham M., Stark H. J., Stammer H., Prätzel-Wunder S., Bickenbach J. R., et al. . (2015). Wnt-3a-activated human fibroblasts promote human keratinocyte proliferation and matrix destruction. Int. J. Cancer 136, 2786–2798. 10.1002/ijc.29336
    1. Spiekstra S. W., Breetveld M., Rustemeyer T., Scheper R., Gibbs S. (2007). Wound-healing factors secreted by epidermal keratinocytes and dermal fibroblasts in skin substitutes. Wound Repair Regen. 15, 708–717. 10.1111/j.1524-475X.2007.00280.x
    1. Tan K. K. B., Salgado G., Connolly J. E., Chan J. K. Y., Lane E. B. (2014). Characterization of fetal keratinocytes, showing enhanced stem cell-like properties: a potential source of cells for skin reconstruction. Stem Cell Rep. 3, 324–338. 10.1016/j.stemcr.2014.06.005
    1. Usui M. L., Mansbridge J. N., Carter W. G., Fujita M., Olerud J. E. (2008). Keratinocyte migration, proliferation, and differentiation in chronic ulcers from patients with diabetes and normal wounds. J. Histochem. Cytochem. 56, 687–696. 10.1369/jhc.2008.951194
    1. Vacanti J. P., Langer R. (1999). Tissue engineering: the design and fabrication of living replacement devices for surgical reconstruction and transplantation. Lancet 354(Suppl. 1), SI32–S134. 10.1016/S0140-6736(99)90247-7
    1. Varkey M., Ding J., Tredget E. E. (2015). Advances in skin substitutes-potential of tissue engineered skin for facilitating anti-fibrotic healing. J. Funct. Biomater. 6, 547–563. 10.3390/jfb6030547
    1. Werner S., Krieg T., Smola H. (2007). Keratinocyte-fibroblast interactions in wound healing. J. Invest. Dermatol. 127, 998–1008. 10.1038/sj.jid.5700786
    1. Wojtowicz A. M., Oliveira S., Carlson M. W., Zawadzka A., Rousseau C. F., Baksh D. (2014). The importance of both fibroblasts and keratinocytes in a bilayered living cellular construct used in wound healing. Wound Repair Regen. 22, 246–255. 10.1111/wrr.12154
    1. Zaja-Milatovic S., Richmond A. (2008). CXC chemokines and their receptors: a case for a significant biological role in cutaneous wound healing. Histol. Histopathol. 23, 1399–1407. 10.14670/HH-23.1399
    1. Zuliani T., Saiagh S., Knol A. C., Esbelin J., Dréno B. (2013). Fetal fibroblasts and keratinocytes with immunosuppressive properties for allogeneic cell-based wound therapy. PLoS ONE 8:e70408. 10.1371/journal.pone.0070408

Source: PubMed

3
Abonnere