Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-c9gpj Total loading time: 0 Render date: 2024-07-12T04:05:41.706Z Has data issue: false hasContentIssue false

Chapter 68 - Regenerative Medicine: The Future?

from Section 17 - Novel Cell Therapies and Manipulations: Ready for Prime-Time?

Published online by Cambridge University Press:  24 May 2017

Hillard M. Lazarus
Affiliation:
Case Western Reserve University, Ohio
Robert Peter Gale
Affiliation:
Imperial College London
Armand Keating
Affiliation:
University of Toronto
Andrea Bacigalupo
Affiliation:
Ospedale San Martino, Genoa
Reinhold Munker
Affiliation:
Louisiana State University, Shreveport
Kerry Atkinson
Affiliation:
University of Queensland
Syed Ali Abutalib
Affiliation:
Midwestern Regional Medical Center, Cancer Treatment Centers of America, Chicago
Get access
Type
Chapter
Information
Hematopoietic Cell Transplants
Concepts, Controversies and Future Directions
, pp. 657 - 673
Publisher: Cambridge University Press
Print publication year: 2000

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Kaiser, L.R.. The future of multihospital systems. Topics in Health Care Financing 1992; 18: 3245.Google ScholarPubMed
McCulloch, E.A. and Till, J.E.. The radiation sensitivity of normal mouse bone marrow cells, determined by quantitative marrow transplantation into irradiated mice. Radiation Research 1960; 13: 115–25.CrossRefGoogle ScholarPubMed
Storek, J. and Witherspoon, R.P.. Immunological reconstitution after hematopoietic stem cell transplantation. In: Atkinson, K., Champlin, R., Ritz, J., Fibbe, W., Ljungman, P. and Brenner, M.K. eds. Clinical Bone Marrow and Blood Stem Cell Transplantation. Cambridge, Cambridge University Press, 2004: 194226.Google Scholar
Kolb, H.J., Atkinson, K. and Reinhold, M.. Cellular therapy. In: The BMT Data Book Including Cellular Therapy. Cambridge, Cambridge University Press, 2013: 209–33.Google Scholar
Weiden, P.L., Flournoy, N., Thomas, E.D. et al. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N Engl J Med. 1979; 300:1068–73.CrossRefGoogle ScholarPubMed
Friedenstein, A.J., Chailakhyan, R.K., Latsinik, N.V. et al. Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo. Transplantation. 1974; 17: 331–40.CrossRefGoogle ScholarPubMed
Barker, N., van de Wetering, M. and Clevers, H.. The intestinal stem cell. Genes Dev. 2008; 22:1856–64.CrossRefGoogle ScholarPubMed
Chen, J.C.J. and Goldhamer, D.J.. Skeletal muscle stem cells. Reprod Biol Endocrinol. 2003; 1: 101–19.Google ScholarPubMed
Blanpain, C. and Fuchs, E.. Epidermal stem cells of the skin. Annu Rev Cell Dev Biol 2006; 22: 339–73.CrossRefGoogle ScholarPubMed
Uchida, N., Buck, D.W., He, S. et al. Direct isolation of human central nervous system stem cells. Proc Nat Acad Sci. 2000; 97: 14720–5.CrossRefGoogle ScholarPubMed
Garget, C.E. and Masuda, H.. Adult stem cells in the endometrium. Mol Hum Reprod. 2010; 16: 818–34.Google Scholar
Beltrami, A.P., Barlucchi, L., Torella, D. et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 2003; 114: 763–76.CrossRefGoogle ScholarPubMed
Myajima, A., Tanaka, M. and Itoh, T.. Stem/progenitor cells in liver development, homeostasis, regeneration, and reprogramming. Cell Stem Cell 2014; 14: 561–74.Google Scholar
Thomson, J.A., Itskovitz-Eldor, J., Sander, S. et al. Embryonic stem cell lines derived from human blastocysts. Science; 1998; 282: 1145–7.CrossRefGoogle ScholarPubMed
Takahashi, K., Tanabe, K., Ohnuki, M. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007; 131: 861–72.CrossRefGoogle ScholarPubMed
Abad, M., Mosteiro, L., Pantoja, C. et al. Reprogramming in vivo produces teratomas and iPS cells with totipotency features. Nature 2013; 502, 340–5.CrossRefGoogle ScholarPubMed
Macchiarini, P., Jungebluth, P., Go, T. et al. Clinical transplantation of a tissue-engineered airway. Lancet; 2008: 372, 2023–30.CrossRefGoogle ScholarPubMed
Sjöqvist, S., Jungebluth, P., Lim, M.L. et al. Experimental orthotopic transplantation of a tissue-engineered oesophagus in rats. Nature Communications 2014, 5: 3562.CrossRefGoogle ScholarPubMed
Atala, A. and Murphy, S.V.. 3D bioprinting of tissues and organs. Nat Biotechnol. 2014; 32: 773–85.Google Scholar
Balsam, L.B., Wagers, A.J., Chistenson, J.L.. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature 2004; 428: 668–73.CrossRefGoogle ScholarPubMed
Saadettin, S., Ulf, M., Schilling, N. et al. Bone marrow cells and CD117-positive haematopoietic stem cells promote corneal wound healing. Acta Ophthalmologica 2012; e367–e373.Google Scholar
Abedi, M., Foster, B.M., Wood, K.D. et al. Haematopoietic stem cells participate in muscle regeneration. Br J Haematol. 2007; 138: 792801.CrossRefGoogle ScholarPubMed
Kovacic, J.C., Macdonald, P., Feneley, M.P. et al. Safety and efficacy of consecutive cycles of granulocyte-colony stimulating factor, and an intracoronary CD133+ cell infusion in patients with chronic refractory ischemic heart disease: The G-CSF in Angina patients with IHD to stimulate Neovascularization (GAIN I) trial. Am Heart J 2008; 156: 954–63.CrossRefGoogle Scholar
Kang, H.-J., Kim, H.-S., Zhang, S.-Y. et al. Effects of intracoronary infusion of peripheral blood stem-cells mobilised with granulocyte-colony stimulating factor on left ventricular systolic function and restenosis after coronary stenting in myocardial infarction: the MAGIC cell randomised clinical trial. Lancet 2004; 363: 751–56.CrossRefGoogle ScholarPubMed
Körbling, M., Katz, R.L., Khanna, A. et al. Hepatocytes and epithelial cells of donor origin in recipients of peripheral-blood stem cells. N Engl J Med. 2002; 346: 738–46.CrossRefGoogle ScholarPubMed
Sacchetti, B., Funari, A., Michienzi, S. et al: Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 2007; 131: 324–36.CrossRefGoogle ScholarPubMed
Mendez-Ferrer, S., Michurina, T.V., Ferraro, F. et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 2010; 466: 829–34.CrossRefGoogle Scholar
de Lima, M., McNiece, I., Robinson, S.N. et al. Cord-blood engraftment with ex-vivo mesenchymal-cell coculture. N Engl J Med. 2012; 367: 2305–15.CrossRefGoogle ScholarPubMed
Crisan, M., Yap, S., Casteilla, L. et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 2008; 3: 301–13.CrossRefGoogle ScholarPubMed
Da Silva Meireles, L., Caplan, A.I., Nardi, N.B. et al. In search of the in vivo identity of mesenchymal stem cells. Stem Cells. 2008; 26: 2287–99.CrossRefGoogle Scholar
Le Blanc, K., Rasmusson, I., Sundberg, B. et al. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 2004; 363: 1439–41.CrossRefGoogle ScholarPubMed
Le Blanc, K., Frassoni, F., Ball, L. et al. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet 2008; 371: 1579–86.CrossRefGoogle ScholarPubMed
Le Blanc, K. and Ringden, O.. Immunobiology of human mesenchymal stem cells and future use in hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2005; 11: 321–34.CrossRefGoogle ScholarPubMed
Singer, N.G. and Caplan, A.I.. Mesenchymal stem cells: mechanisms of inflammation. Annu Rev Pathol Mech Dis. 2011; 6: 457–78.CrossRefGoogle ScholarPubMed
Pelekanos, R., Gongora, L.I., Doan, T. et al. Comprehensive genotype/phenotype analysis of renal and cardiac MSC-like populations supports strong congruence with bone marrow MSC despite maintenance of distinct identities. Stem Cell Res. 2012; 5: 5873.Google Scholar
Barlow, S., Brooke, G., Chatterjee, K. et al. Comparison of human placenta- and bone marrow-derived multipotent mesenchymal stem cells. Stem Cells Dev 2008; 17: 1095–108.CrossRefGoogle ScholarPubMed
Maxson, S., Lopez, E.A., Yoo, D. et al. Concise Review: Role of mesenchymal stem cells in wound repair. Stem Cells Transl Med. 2012; 1: 142–9.CrossRefGoogle ScholarPubMed
Badivas, E.V., Abedi, M., Butmarc, J. et al. Participation of bone marrow derived cells in cutaneous wound healing. J Cell Physiol. 2003; 196: 245–50.Google Scholar
Falanga, V., Iwamoto, S., Chartier, M. et al. Autologous bone marrow-derived cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing in murine and human cutaneous wounds. Tissue Eng. 2007; 13: 1299–312.CrossRefGoogle Scholar
Yoshikawa, T., Mitsuno, H., Nonaka, I. et al. Wound therapy by marrow mesenchymal cell transplantation. Plast Reconstr Surg. 2008; 121: 860–77.CrossRefGoogle ScholarPubMed
Lu, D., Chen, B. and Liang, Z.. Comparison of bone marrow mesenchymal stem cells with bone marrow-derived mononuclear cells for treatment of diabetic critical limb ischemia and foot ulcer: a double-blind, randomized, controlled trial. Diabetes Res Clin Pract. 2011; 92: 2636.CrossRefGoogle ScholarPubMed
Lee, H.C., An, S.G., Lee, H.W. et al. Safety and effect of adipose tissue-derived stem cell implantation in patients with critical limb ischemia: a pilot study. Circ J. 2012; 76: 1750–60.CrossRefGoogle ScholarPubMed
Garcia-Olmo, D., Garcia-Arranz, M. and Herreros, D.. Expanded adipose-derived stem cells for the treatment of complex perianal fistula including Crohn’s disease. Expert Opin Biol Ther. 2008; 8: 1417–23.CrossRefGoogle ScholarPubMed
García-Olmo, D., Herreros, D., De-La-Quintana, P. et al. Adipose-derived stem cells in Crohn’s rectovaginal fistula. Case Rep Med. 2010: 961758.CrossRefGoogle Scholar
de Steiger, R., Ferrugia, R, Richardson, M. et al. OR14: A safety study of autologous mesenchymal precursor cells in the management of non-union of tibial and fibular fractures. J Bone Joint Surg Br. 2010; 92: 203–4.Google Scholar
Quarto, R., Mastrogiacomo, M., Cancedda, R. et al. Repair of large bone defects with the use of autologous bone marrow stromal cells. N Engl J Med. 2001; 344: 385–6.CrossRefGoogle ScholarPubMed
Gimbel, M., Ashley, R.K., Sisodia, M. et al. Repair of alveolar cleft defects: reduced morbidity with bone marrow stem cells in a resorbable matrix. J Craniofac Surg. 2007; 18: 895901.CrossRefGoogle Scholar
Velardi, F., Amante, P.R., Caniglia, M. et al. Osteogenesis induced by autologous bone marrow cells transplant in the pediatric skull. Childs Nerv Syst. 2006; 22: 1158–66.CrossRefGoogle ScholarPubMed
Gan, Y., Dai, K., Zhang, P. et al. The clinical use of enriched bone marrow stem cells combined with porous beta-tricalcium phosphate in posterior spinal fusion. Biomaterials 2008; 29: 3973–82.CrossRefGoogle ScholarPubMed
Vangsness, C.T., David, T.D., David, W.G. et al. A randomized clinical trial using mesenchymal stem cells for meniscus regeneration and osteoarthritis. http://www.aaos.org/Education/anmeet/education/Sports_Medicine_Arthoroscopy_Abstracts.pdf 2012.Google Scholar
Nejadnik, H., Hui, J.H., Feng Choong, E.P. et al. Autologous bone marrow-derived mesenchymal stem cells versus autologous chondrocyte implantation: an observational cohort study. Am J Sports Med. 2010; 38: 1110–16.CrossRefGoogle ScholarPubMed
Wakitani, S., Nawata, M., Tensho, K. et al. Repair of articular cartilage defects in the patello-femoral joint with autologous bone marrow mesenchymal cell transplantation: three case reports involving nine defects in five knees. J Tissue Eng Regener Med 2007; 1: 74–9.CrossRefGoogle ScholarPubMed
Kuroda, R., Ishida, K., Matsumoto, T. et al. Treatment of a full-thickness articular cartilage defect in the femoral condyle of an athlete with autologous bone-marrow stromal cells. Osteoarthritis Cartilage 2007; 15: 226–31.CrossRefGoogle ScholarPubMed
Da Silva, J.S. and Hare, J.M.. Cell-based therapies for myocardial repair: emerging role for bone marrow-derived mesenchymal stem cells (MSCs) in the treatment of the chronically injured heart. Methods Mol Biol. 2013; 1037: 145–63.CrossRefGoogle ScholarPubMed
Heldman, A.W., DiFede, D.L., Fishman, J.E. et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC-HFT randomized trial. JAMA. 2014; 311: 6273.CrossRefGoogle ScholarPubMed
Loffredo, F.S., Steinhauser, M.L., Gannon, J. et al. Bone marrow-derived cell therapy stimulates endogenous cardiomyocyte progenitors and promotes cardiac repair. Cell Stem Cell 2011; 8: 389–98.CrossRefGoogle ScholarPubMed
Chambers, D.C., Enever, D., Ilic, N. et al. Safety of mesenchymal stromal cell therapy for idiopathic pulmonary fibrosis. Respirology. In press.Google Scholar
Sun, L., Wang, D., Liang, J. et al. Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus. Arthritis Rheum. 2010; 62: 2467–75.CrossRefGoogle ScholarPubMed
Liang, J., Zhang, H. and Hua, B.. Allogeneic mesenchymal stem cells transplantation in refractory systemic lupus erythematosus: a pilot clinical study. Ann Rheum Dis. 2010; 69: 1423–9.CrossRefGoogle ScholarPubMed
Yamout, B., Hourani, R., Salti, H. et al. Bone marrow mesenchymal stem cell transplantation in patients with multiple sclerosis: a pilot study. J Neuroimmunol. 2010; 227: 185–9.CrossRefGoogle ScholarPubMed
Makkar, R.R., Smith, R.R., Cheng, K. et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase I trial. Lancet. 2012; 379: 895904.CrossRefGoogle Scholar
Malliaras, K., Makkar, R.R., Smith, R.R. et al. Intracoronary cardiosphere-derived cells after myocardial infarction: evidence of therapeutic regeneration in the final 1-year results of the CADUCEUS trial (CArdiosphere-Derived aUtologous stem CElls to reverse ventricUlar dySfunction). J Am Coll Cardiol. 2014; 63: 110–22.CrossRefGoogle ScholarPubMed
Menache, P.. Skeletal myoblasts and cardiac repair. J Mol Cell Cardiol. 2008; 45: 545–53.Google Scholar
Uchida, N., Buck, D.W., He, S. et al. Direct isolation of human central nervous system stem cells. Proc Nat Acad Sci USA. 2000; 97: 14720–5.CrossRefGoogle ScholarPubMed
Tsukamoto, A., Uchida, N., Capela, A. et al. Clinical transplantation of human neural stem cells. Stem Cell Res Ther. 2013; 4: 102–13.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×