Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-sxzjt Total loading time: 0 Render date: 2024-04-18T15:45:30.968Z Has data issue: false hasContentIssue false

Chapter 10 - CD34+ Cell Mobilization in Autologous and Allogeneic Hematopoietic Cell Transplants

from Section 3 - Collecting and Processing of the Graft

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. 79 - 96
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

Bensinger, WI, Martin, PJ, Storer, B, Clift, R, Forman, SJ, Negrin, R, et al. Transplantation of bone marrow as compared with peripheral-blood cells from HLA-identical relatives in patients with hematologic cancers. N Engl J Med. 2001; 344(3):175–81.Google Scholar
Beyer, J, Schwella, N, Zingsem, J, Strohscheer, I, Schwaner, I, Oettle, H, et al. Hematopoietic rescue after high-dose chemotherapy using autologous peripheral-blood progenitor cells or bone marrow: a randomized comparison. J Clin Oncol. 1995; 13(6):1328–35.Google Scholar
Hartmann, O, Le Corroller, AG, Blaise, D, Michon, J, Phillip, I, Norol, F, et al. Peripheral blood stem cell and bone marrow transplantation for solid tumors and lymphomas: hematologic recovery and costs. A randomized, controlled trial. Ann Intern Med. 1997; 126(8):600–7.Google Scholar
Schmitz, N, Linch, DC, Dreger, P, Goldstone, AH, Boogaerts, MA, Ferrant, A, et al. Randomised trial of filgrastim-mobilised peripheral blood progenitor cell transplantation versus autologous bone-marrow transplantation in lymphoma patients. Lancet. 1996; 347(8998):353–7.CrossRefGoogle ScholarPubMed
Bender, JG, To, LB, Williams, S, Schwartzberg, LS. Defining a therapeutic dose of peripheral blood stem cells. J Hematother. 1992; 1(4):329–41.Google Scholar
Passos-Coelho, JL, Braine, HG, Davis, JM, Huelskamp, AM, Schepers, KG, Ohly, K, et al. Predictive factors for peripheral-blood progenitor-cell collections using a single large-volume leukapheresis after cyclophosphamide and granulocyte-macrophage colony-stimulating factor mobilization. J Clin Oncol. 1995; 13(3):705–14.Google Scholar
Gandhi, MK, Jestice, K, Scott, MA, Bloxham, D, Bass, G, Marcus, RE. The minimum CD34 threshold depends on prior chemotherapy in autologous peripheral blood stem cell recipients. Bone Marrow Transplant. 1999; 23(1):913.Google Scholar
Montgomery, M, Cottler-Fox, M. Mobilization and collection of autologous hematopoietic progenitor/stem cells. Clin Adv Hematol Oncol. 2007; 5(2):127–36.Google Scholar
Weaver, CH, Hazelton, B, Birch, R, Palmer, P, Allen, C, Schwartzberg, L, et al. An analysis of engraftment kinetics as a function of the CD34 content of peripheral blood progenitor cell collections in 692 patients after the administration of myeloablative chemotherapy. Blood. 1995; 86(10):3961–9.Google Scholar
Beguin, Y, Baudoux, E, Sautois, B, Fraipont, V, Schaaf-Lafontaine, N, Pereira, M, et al. Hematopoietic recovery in cancer patients after transplantation of autologous peripheral blood CD34+ cells or unmanipulated peripheral blood stem and progenitor cells. Transfusion. 1998; 38(2):199208.Google Scholar
Reiffers, J, Faberes, C, Boiron, JM, Marit, G, Foures, C, Ferrer, AM, et al. Peripheral blood progenitor cell transplantation in 118 patients with hematological malignancies: analysis of factors affecting the rate of engraftment. J Hematother. 1994; 3(3):185–91.Google Scholar
Bolwell, BJ, Pohlman, B, Rybicki, L, Sobecks, R, Dean, R, Curtis, J, et al. Patients mobilizing large numbers of CD34+ cells (‘super mobilizers’) have improved survival in autologous stem cell transplantation for lymphoid malignancies. Bone Marrow Transplant. 2007; 40(5):437–41.Google Scholar
Pulsipher, MA, Chitphakdithai, P, Logan, BR, Leitman, SF, Anderlini, P, Klein, JP, et al. Donor, recipient, and transplant characteristics as risk factors after unrelated donor PBSC transplantation: beneficial effects of higher CD34+ cell dose. Blood. 2009; 114(13):2606–16.Google Scholar
Baron, F, Maris, MB, Storer, BE, Sandmaier, BM, Panse, JP, Chauncey, TR, et al. High doses of transplanted CD34+ cells are associated with rapid T-cell engraftment and lessened risk of graft rejection, but not more graft-versus-host disease after nonmyeloablative conditioning and unrelated hematopoietic cell transplantation. Leukemia. 2005; 19(5):822–8.Google Scholar
Dreger, P, Kloss, M, Petersen, B, Haferlach, T, Loffler, H, Loeffler, M, et al. Autologous progenitor cell transplantation: prior exposure to stem cell-toxic drugs determines yield and engraftment of peripheral blood progenitor cell but not of bone marrow grafts. Blood. 1995; 86(10):3970–8.Google Scholar
Tarella, C, Di Nicola, M, Caracciolo, D, Zallio, F, Cuttica, A, Omede, P. High-dose ara-C with autologous peripheral blood progenitor cell support induces a marked progenitor cell mobilization: an indication for patients at risk for low mobilization. Bone Marrow Transplant. 2002; 30(11):725–32.Google Scholar
Pusic, I, Jiang, SY, Landua, S, Uy, GL, Rettig, MP, Cashen, AF, et al. Impact of mobilization and remobilization strategies on achieving sufficient stem cell yields for autologous transplantation. Biol Blood Marrow Transplant. 2008; 14(9): 1045–56.CrossRefGoogle ScholarPubMed
Fadini, GP, Pucci, L, Vanacore, R, Baesso, I, Penno, G, Balbarini, A, et al. Glucose tolerance is negatively associated with circulating progenitor cell levels. Diabetologia. 2007; 50(10):2156–63.CrossRefGoogle ScholarPubMed
Fadini, GP, Boscaro, E, de Kreutzenberg, S, Agostini, C, Seeger, F, Dimmeler, S, et al. Time course and mechanisms of circulating progenitor cell reduction in the natural history of type 2 diabetes. Diabetes Care. 2010; 33(5):1097–102.Google Scholar
Ferraro, F, Lymperi, S, Mendez-Ferrer, S, Saez, B, Spencer, JA, Yeap, BY, et al. Diabetes impairs hematopoietic stem cell mobilization through alteration of niche function. Sci Transl Med. 2011; 3(104):104ra101.CrossRefGoogle ScholarPubMed
Heazlewood, SY, Oteiza, A, Cao, H, Nilsson, SK. Analyzing hematopoietic stem cell homing, lodgment and engraftment to better understand the bone marrow niche. Ann N Y Acad Sci. 2014; 1310:119–28.Google Scholar
Rettig, MP, Schroeder, MA, DiPersio, JF. Marrow microenvironment and biology of mobilization of stem cells. In: Appelbaum, FR, Forman, SJ, Negrin, RS, Blume, KG, editors. Thomas’ Hematopoietic Cell Transplantation, 5th edition. New Jersey: Wiley-Blackwell; 2013.Google Scholar
Morrison, SJ, Scadden, DT. The bone marrow niche for haematopoietic stem cells. Nature. 2014; 505(7483):327–34.Google Scholar
Taichman, RS, Emerson, SG. Human osteoblasts support hematopoiesis through the production of granulocyte colony-stimulating factor. J Exp Med. 1994; 179(5):1677–82.Google Scholar
Calvi, LM, Adams, GB, Weibrecht, KW, Weber, JM, Olson, DP, Knight, MC, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003; 425:841–6.Google Scholar
Park, D, Spencer, JA, Koh, BI, Kobayashi, T, Fujisaki, J, Clemens, TL, et al. Endogenous bone marrow MSCs are dynamic, fate-restricted participants in bone maintenance and regeneration. Cell Stem Cell. 2012; 10(3):259–72.Google Scholar
Zhang, J, Niu, C, Ye, L, Huang, H, He, X, Tong, WG, et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003; 425(6960):836–41.CrossRefGoogle ScholarPubMed
Kiel, MJ, Yilmaz, OH, Iwashita, T, Yilmaz, OH, Terhorst, C, Morrison, SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005; 121(7):1109–21.Google Scholar
Kiel, MJ, Radice, GL, Morrison, SJ. Lack of evidence that hematopoietic stem cells depend on N-cadherin-mediated adhesion to osteoblasts for their maintenance. Cell Stem Cell. 2007; 1(2):204–17.Google Scholar
Visnjic, D, Kalajzic, Z, Rowe, DW, Katavic, V, Lorenzo, J, Aquila, HL. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood. 2004; 103(9):3258–64.CrossRefGoogle ScholarPubMed
Zhu, J, Garrett, R, Jung, Y, Zhang, Y, Kim, N, Wang, J, et al. Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells. Blood. 2007; 109(9):3706–12.Google Scholar
Raaijmakers, MH, Mukherjee, S, Guo, S, Zhang, S, Kobayashi, T, Schoonmaker, JA, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukemia. Nature. 2010; 464(7290):852–7.CrossRefGoogle Scholar
Kiel, MJ, Acar, M, Radice, GL, Morrison, SJ. Hematopoietic stem cells do not depend on N-cadherin to regulate their maintenance. Cell Stem Cell. 2009; 4(2):170–9.CrossRefGoogle Scholar
Greenbaum, AM, Revollo, LD, Woloszynek, JR, Civitelli, R, Link, DC. N-cadherin in osteolineage cells is not required for maintenance of hematopoietic stem cells. Blood. 2012; 120(2):295302.Google Scholar
Bromberg, O, Frisch, BJ, Weber, JM, Porter, RL, Civitelli, R, Calvi, LM. Osteoblastic N-cadherin is not required for microenvironmental support and regulation of hematopoietic stem and progenitor cells. Blood. 2012; 120(2):303–13.Google Scholar
Zhou, X, Zhang, Z, Feng, JQ, Dusevich, VM, Sinha, K, Zhang, H, et al. Multiple functions of Osterix are required for bone growth and homeostasis in postnatal mice. Proc Natl Acad Sci USA. 2010; 107(29):12919–24.Google Scholar
Chan, CK, Chen, CC, Luppen, CA, Kim, JB, DeBoer, AT, Wei, K, et al. Endochondral ossification is required for haematopoietic stem-cell niche formation. Nature. 2009; 457:490–4.Google Scholar
Sacchetti, B, Funari, A, Michienzi, S, Di Cesare, S, Piersanti, S, Saggio, I, et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell. 2007; 131(2):324–36.Google Scholar
Sugiyama, T, Kohara, H, Noda, M, Nagasawa, T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. 2006; 25(6):977–88.Google Scholar
Lo, CC, Fleming, HE, Wu, JW, Zhao, CX, Miake-Lye, S, Fujisaki, J, et al. Live-animal trafficking of individual haematopoietic stem/progenitor cells in their niche. Nature. 2009; 457(7225):92–6.Google Scholar
Sipkins, DA, Wei, X, Wu, JW, Runnels, JM, Cote, D, Means, TK, et al. In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature. 2005; 435(7044):969–73.Google Scholar
Mendez-Ferrer, S, Michurina, TV, Ferraro, F, Mazloom, AR, MacArthur, BD, Lira, SA, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010; 466:829–34.Google Scholar
Yao, L, Yokota, T, Xia, L, Kincade, PW, McEver, RP. Bone marrow dysfunction in mice lacking the cytokine receptor gp130 in endothelial cells. Blood. 2005; 106(13):4093–101.Google Scholar
Hooper, AT, Butler, JM, Nolan, DJ, Kranz, A, Iida, K, Kobayashi, M, et al. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell. 2009; 4(3):263–74.Google Scholar
Oguro, H, Ding, L, Morrison, SJ. SLAM family markers resolve functionally distinct subpopulations of hematopoietic stem cells and multipotent progenitors. Cell Stem Cell. 2013; 13(1):102–16.Google Scholar
Barker, JE. Early transplantation to a normal microenvironment prevents the development of Steel hematopoietic stem cell defects. Exp Hematol. 1997; 25(6):542–7.Google ScholarPubMed
Tzeng, YS, Li, H, Kang, YL, Chen, WC, Cheng, WC, Lai, DM. Loss of Cxcl12/Sdf-1 in adult mice decreases the quiescent state of hematopoietic stem/progenitor cells and alters the pattern of hematopoietic regeneration after myelosuppression. Blood. 2011; 117(2):429–39.Google Scholar
Zou, YR, Kottmann, AH, Kuroda, M, Taniuchi, I, Littman, DR. Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature. 1998; 393(6685):595–9.Google Scholar
Katayama, Y, Battista, M, Kao, WM, Hidalgo, A, Peired, AJ, Thomas, SA, et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006; 124(2):407–21.Google Scholar
Mendez-Ferrer, S, Lucas, D, Battista, M, Frenette, PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008; 452(7186):442–7.Google Scholar
Katayama, Y, Battista, M, Kao, WM, Hidalgo, A, Peired, AJ, Thomas, SA, et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006; 124(2):407–21.CrossRefGoogle ScholarPubMed
Himburg, HA, Muramoto, GG, Daher, P, Meadows, SK, Russell, JL, Doan, P, et al. Pleiotrophin regulates the expansion and regeneration of hematopoietic stem cells. Nat Med. 2010; 16(4):475–82.Google Scholar
Himburg, HA, Harris, JR, Ito, T, Daher, P, Russell, JL, Quarmyne, M, et al. Pleiotrophin regulates the retention and self-renewal of hematopoietic stem cells in the bone marrow vascular niche. Cell Rep. 2012; 2(4):964–75.Google Scholar
Petit, I, Szyper-Kravitz, M, Nagler, A, Lahav, M, Peled, A, Habler, L, et al. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol. 2002; 3(7):687–94.Google Scholar
Nakamura-Ishizu, A, Okuno, Y, Omatsu, Y, Okabe, K, Morimoto, J, Uede, T, et al. Extracellular matrix protein tenascin-C is required in the bone marrow microenvironment primed for hematopoietic regeneration. Blood. 2012; 119(23):5429–37.Google Scholar
Stier, S, Ko, Y, Forkert, R, Lutz, C, Neuhaus, T, Grunewald, E, et al. Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size. J Exp Med. 2005; 201(11):1781–91.Google Scholar
Nilsson, SK, Johnston, HM, Whitty, GA, Williams, B, Webb, RJ, Denhardt, DT, et al. Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood. 2005; 106(4):1232–9.Google Scholar
Sugimura, R, He, XC, Venkatraman, A, Arai, F, Box, A, Semerad, C, et al. Noncanonical Wnt signaling maintains hematopoietic stem cells in the niche. Cell. 2012; 150(2):351–65.Google Scholar
Christopher, MJ, Rao, M, Liu, F, Woloszynek, JR, Link, DC. Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. J Exp Med. 2011; 208(2):251–60.Google Scholar
Liu, F, Poursine-Laurent, J, Link, DC. Expression of the G-CSF receptor on hematopoietic progenitor cells is not required for their mobilization by G-CSF. Blood. 2000; 95(10):3025–31.Google Scholar
Anderlini, P, Przepiorka, D, Seong, D, Miller, P, Sundberg, J, Lichtiger, B, et al. Clinical toxicity and laboratory effects of granulocyte-colony-stimulating factor (filgrastim) mobilization and blood stem cell apheresis from normal donors, and analysis of chargers for the procedures. Transfusion. 1996; 36(7):590–5.Google Scholar
Stroncek, DF, Clay, ME, Petzoldt, ML, Smith, J, Jaszcz, W, Oldham, FB, et al. Treatment of normal individuals with granulocyte-colony-stimulating factor: donor experiences and the effects on peripheral blood CD34+ cell counts and on the collection of peripheral blood stem cells. Transfusion. 1996; 36(7):601–10.Google Scholar
Becker, PS, Wagle, M, Matous, S, Swanson, RS, Pihan, G, Lowry, PA, et al. Spontaneous splenic rupture following administration of granulocyte colony-stimulating factor (G-CSF): occurrence in an allogeneic donor of peripheral blood stem cells. Biol Blood Marrow Transplant. 1997; 3(1):45–9.Google Scholar
Stroncek, D, Shawker, T, Follmann, D, Leitman, SF. G-CSF-induced spleen size changes in peripheral blood progenitor cell donors. Transfusion. 2003; 43(5):609–13.CrossRefGoogle ScholarPubMed
Rothe, L, Collin-Osdoby, P, Chen, Y, Sunyer, T, Chaudhary, L, Tsay, A, et al. Human osteoclasts and osteoclast-like cells synthesize and release high basal and inflammatory stimulated levels of the potent chemokine interleukin-8. Endocrinology. 1998; 139(10):4353–63.CrossRefGoogle ScholarPubMed
Korbling, M, Fliender, TM. History of blood stem cell transplants. Blood stem cell transplants. In: Gale, RP, Juttner, CA, Henon, P, editors. Peripheral blood stem cell autographs. New York: Cambridge University Press; 1994:9.Google Scholar
Blaise, D, Kuentz, M, Fortanier, C, Bourhis, JH, Milpied, N, Sutton, L, et al. Randomized trial of bone marrow versus lenograstim-primed blood cell allogeneic transplantation in patients with early-stage leukemia: a report from the Societe Francaise de Greffe de Moelle. J Clin Oncol. 2000; 18(3):537–46.Google Scholar
Couban, S, Simpson, DR, Barnett, MJ, Bredeson, C, Hubesch, L, Howson-Jan, K, et al. A randomized multicenter comparison of bone marrow and peripheral blood in recipients of matched sibling allogeneic transplants for myeloid malignancies. Blood. 2002; 100(5):1525–31.Google Scholar
Schmitz, N, Beksac, M, Hasenclever, D, Bacigalupo, A, Ruutu, T, Nagler, A, et al. Transplantation of mobilized peripheral blood cells to HLA-identical siblings with standard-risk leukemia. Blood. 2002; 100(3):761–7.Google Scholar
Goterris, R, Hernandez-Boluda, JC, Teruel, A, Gomez, C, Lis, MJ, Terol, MJ, et al. Impact of different strategies of second-line stem cell harvest on the outcome of autologous transplantation in poor peripheral blood stem cell mobilizers. Bone Marrow Transplant. 2005; 36(10):847–53.Google Scholar
Costa, LJ, Kramer, C, Hogan, KR, Butcher, CD, Littleton, AL, Shoptaw, KB, et al. Pegfilgrastim-versus filgrastim-based autologous hematopoietic stem cell mobilization in the setting of preemptive use of plerixafor: efficacy and cost analysis. Transfusion. 2012; 52(11):2375–81.Google Scholar
Herbert, KE, Gambell, P, Link, EK, Mouminoglu, A, Wall, DM, Harrison, SJ, et al. Pegfilgrastim compared with filgrastim for cytokine-alone mobilization of autologous haematopoietic stem and progenitor cells. Bone Marrow Transplant. 2013; 48(3):351–6.CrossRefGoogle ScholarPubMed
Bruns, I, Steidl, U, Fischer, JC, Raschke, S, Kobbe, G, Fenk, R, et al. Pegylated G-CSF mobilizes CD34+ cells with different stem and progenitor cell subsets and distinct functional properties in comparison with unconjugated G-CSF. Blood (ASH Annual Meeting Abstracts). 2006; 108:Abstract 3382.Google Scholar
Kobbe, G, Bruns, I, Fenk, R, Czibere, A, Haas, R. Pegfilgrastim for PBSC mobilization and autologous haematopoietic SCT. Bone Marrow Transplant. 2009; 43(9):669–77.Google Scholar
Sarkar, CA, Lowenhaupt, K, Wang, PJ, Horan, T, Lauffenburger, DA. Parsing the effects of binding, signaling, and trafficking on the mitogenic potencies of granulocyte colony-stimulating factor analogues. Biotechnol Prog. 2003; 19(3):955–64.Google Scholar
Morris, ES, MacDonald, KP, Hill, GR. Stem cell mobilization with G-CSF analogs: a rational approach to separate GVHD and GVL? Blood. 2006; 107(9):3430–5.CrossRefGoogle ScholarPubMed
Brown, RA, Adkins, D, Khoury, H, Vij, R, Goodnough, LT, Shenoy, S, et al. Long-term follow-up of high-risk allogeneic peripheral-blood stem-cell transplant recipients: graft-versus-host disease and transplant-related mortality. J Clin Oncol. 1999; 17(3):806–12.Google Scholar
Lane, TA, Law, P, Maruyama, M, Young, D, Burgess, J, Mullen, M, et al. Harvesting and enrichment of hematopoietic progenitor cells mobilized into the peripheral blood of normal donors by granulocyte-macrophage colony-stimulating factor (GM-CSF) or G-CSF: potential role in allogeneic marrow transplantation. Blood. 1995; 85(1):275–82.Google Scholar
Gazitt, Y, Shaughnessy, P, Liu, Q. Differential mobilization of CD34+ cells and lymphoma cells in non-Hodgkin’s lymphoma patients mobilized with different growth factors. J Hematother Stem Cell Res. 2001; 10(1):167–76.Google ScholarPubMed
Devine, SM, Brown, RA, Mathews, V, Trinkaus, K, Khoury, H, Adkins, D, et al. Reduced risk of acute GvHD following mobilization of HLA-identical sibling donors with GM-CSF alone. Bone Marrow Transplant. 2005; 36(6):531–8.Google Scholar
Gazitt, Y, Shaughnessy, P, Devore, P. Mobilization of dendritic cells and NK cells in non-Hodgkin’s lymphoma patients mobilized with different growth factors. J Hematother Stem Cell Res. 2001; 10(1):177–86.Google Scholar
Schroeder, MA, Merida, S, Schwab, D, Rettig, MP, Meier, S, Lopez, S, et al. Sargramostim (GM-CSF) combined with IV Plerixafor to mobilize peripheral blood stem cells (PBSC) from normal HLA-matched allogeneic sibling donors. BMT Tandem “Scientific” Meeting, March, 2014; Session N(Abstract 32):Abstract 32.Google Scholar
Ford, CD, Greenwood, J, Anderson, J, Snow, G, Petersen, FB. CD34+ cell adhesion molecule profiles differ between patients mobilized with granulocyte-colony-stimulating factor alone and chemotherapy followed by granulocyte-colony-stimulating factor. Transfusion. 2006; 46(2):193–8.Google Scholar
Milone, G, Leotta, S, Indelicato, F, Mercurio, S, Moschetti, G, Di Raimondo, F, et al. G-CSF alone vs cyclophosphamide plus G-CSF in PBPC mobilization of patients with lymphoma: results depend on degree of previous treatment. Bone Marrow Transplant. 2003; 31(9):747–54.Google Scholar
Krishnan, A, Bhatia, S, Slovak, ML, Arber, DA, Niland, JC, Nademanee, A, et al. Predictors of therapy-related leukemia and myelodysplasia following autologous transplantation for lymphoma: an assessment of risk factors. Blood. 2000; 95(5):1588–93.Google Scholar
Desikan, KR, Barlogie, B, Jagannath, S, Vesole, DH, Siegel, D, Fassas, A, et al. Comparable engraftment kinetics following peripheral-blood stem-cell infusion mobilized with granulocyte colony-stimulating factor with or without cyclophosphamide in multiple myeloma. J Clin Oncol. 1998; 16(4):1547–53.Google Scholar
Koc, ON, Gerson, SL, Cooper, BW, Laughlin, M, Meyerson, H, Kutteh, L, et al. Randomized cross-over trial of progenitor-cell mobilization: high-dose cyclophosphamide plus granulocyte colony-stimulating factor (G-CSF) versus granulocyte-macrophage colony-stimulating factor plus G-CSF. J Clin Oncol. 2000; 18(9):1824–30.Google Scholar
Gupta, S, Zhou, P, Hassoun, H, Kewalramani, T, Reich, L, Costello, S, et al. Hematopoietic stem cell mobilization with intravenous melphalan and G-CSF in patients with chemoresponsive multiple myeloma: report of a phase II trial. Bone Marrow Transplant. 2005; 35(5):441–7.Google Scholar
Hopman, RK, DiPersio, JF. Advances in stem cell mobilization. Blood Rev. 2014; 28(1):3140.Google Scholar
Ding, L, Saunders, TL, Enikolopov, G, Morrison, SJ. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature. 2012; 481(7382):457–62.Google Scholar
Broudy, VC. Stem cell factor and hematopoiesis. Blood. 1997; 90(4):1345–64.Google Scholar
Levesque, JP, Leavesley, DI, Niutta, S, Vadas, M, Simmons, PJ. Cytokines increase human hematopoietic cell adhesiveness by activation of very late antigen (VLA)-4 and VLA-5 integrins. J Exp Med. 1995; 181(5):1805–15.Google Scholar
da Silva, MG, Pimentel, P, Carvalhais, A, Barbosa, I, Machado, A, Campilho, F,et al. Ancestim (recombinant human stem cell factor, SCF) in association with filgrastim does not enhance chemotherapy and/or growth factor-induced peripheral blood progenitor cell (PBPC) mobilization in patients with a prior insufficient PBPC collection. Bone Marrow Transplant. 2004; 34(8):683–91.Google Scholar
To, LB, Bashford, J, Durrant, S, MacMillan, J, Schwarer, AP, Prince, HM, et al. Successful mobilization of peripheral blood stem cells after addition of ancestim (stem cell factor) in patients who had failed a prior mobilization with filgrastim (granulocyte colony-stimulating factor) alone or with chemotherapy plus filgrastim. Bone Marrow Transplant. 2003; 31(5):371–8.Google Scholar
Lapierre, V, Rossi, JF, Heshmati, F, Azar, N, Vekhof, A, Makowski, C, et al. Ancestim (r-metHuSCF) plus filgrastim and/or chemotherapy for mobilization of blood progenitors in 513 poorly mobilizing cancer patients: the French compassionate experience. Bone Marrow Transplant. 2011; 46(7):936–42.Google Scholar
De Clercq, E. The bicyclam AMD3100 story. Nat Rev Drug Discov. 2003; 2(7):581–7.Google Scholar
Donzella, GA, Schols, D, Lin, SW, Este, JA, Nagashima, KA, Maddon, PJ, et al. AMD3100, a small molecule inhibitor of HIV-1 entry via the CXCR4 co-receptor. Nat Med. 1998; 4(1):72–7.Google Scholar
Devine, SM, Flomenberg, N, Vesole, DH, Liesveld, J, Weisdorf, D, Badel, K, et al. Rapid mobilization of CD34+ cells following administration of the CXCR4 antagonist AMD3100 to patients with multiple myeloma and non-Hodgkin’s lymphoma. J Clin Oncol. 2004; 22(6):1095–102.CrossRefGoogle ScholarPubMed
Flomenberg, N, Devine, SM, DiPersio, JF, Liesveld, JL, McCarty, JM, Rowley, SD, et al. The use of AMD3100 plus G-CSF for autologous hematopoietic progenitor cell mobilization is superior to G-CSF alone. Blood. 2005; 106(5):1867–74.Google Scholar
Calandra, G, McCarty, J, McGuirk, J, Tricot, G, Crocker, SA, Badel, K, et al. AMD3100 plus G-CSF can successful mobilize CD34+ cells from non-Hodgkin’s lymphoma, Hodgkin’s disease and multiple myeloma patients previously failing mobilization with chemotherapy and/or cytokine treatment: compassionate use data. Bone Marrow Transplant. 2008; 41(4):331–8.Google Scholar
Cashen, A, Lopez, S, Gao, F, Calandra, G, MacFarland, R, Badel, K, et al. A phase II study of plerixafor (AMD3100) plus G-CSF for autologous hematopoietic progenitor cell mobilization in patients with Hodgkin lymphoma. Biol Blood Marrow Transplant. 2008; 14(11):1253–61.Google Scholar
Fowler, CJ, Dunn, A, Hayes-Lattin, B, Hansen, K, Hansen, L, Lanier, K, et al. Rescue from failed growth factor and/or chemotherapy HSC mobilization with G-CSF and plerixafor (AMD3100): an institutional experience. Bone Marrow Transplant. 209; 43(12):9–17.Google Scholar
Stiff, P, Micallef, I, McCarthy, P, Magalhaes-Silverman, M, Weisdorf, D, Territo, M, et al. Treatment with plerixafor in non-Hodgkin’s lymphoma and multiple myeloma patients to increase the number of peripheral blood stem cells when given a mobilizing regimen of G-CSF: implications for the heavily pretreated patient. Biol Blood Marrow Transplant. 2009; 15(2):249–56.Google Scholar
Dugan, MJ, Maziarz, RT, Bensinger, WI, Nademanee, A, Liesveld, J, Badel, K, et al. Safety and preliminary efficacy of plerixafor (Mozobil) in combination with chemotherapy and G-CSF: an open-label, multicenter, exploratory trial in patients with multiple myeloma and non-Hodgkin’s lymphoma undergoing stem cell mobilization. Bone Marrow Transplant. 2010; 45(1):3947.Google Scholar
DiPersio, JF, Micallef, IN, Stiff, PJ, Bolwell, BJ, Maziarz, RT, Jacobsen, E, et al. Phase III prospective randomized double-blind placebo-controlled trial of plerixafor plus granulocyte colony-stimulating factor compared with placebo plus granulocyte colony-stimulating factor for autologous stem-cell mobilization and transplantation for patients with non-Hodgkin’s lymphoma. J Clin Oncol. 2009; 27(28):4767–73.Google Scholar
DiPersio, JF, Stadtmauer, EA, Nademanee, A, Micallef, IN, Stiff, PJ, Kaufman, JL, et al. Plerixafor and G-CSF versus placebo and G-CSF to mobilize hematopoietic stem cells for autologous stem cell transplantation in patients with multiple myeloma. Blood. 2009; 113(23):5720–6.Google Scholar
Devine, SM, Vij, R, Rettig, M, Todt, L, McGlauchlen, K, Fisher, N, et al. Rapid mobilization of functional donor hematopoietic cells without G-CSF using AMD3100, an antagonist of the CXCR4/SDF-1 interaction. Blood. 2008; 112(4):990–8.Google Scholar
Rettig, MP, Ansstas, G, DiPersio, JF. Mobilization of hematopoietic stem and progenitor cells using inhibitors of CXCR4 and VLA-4. Leukemia. 2012; 26(1):3453.Google Scholar
Nazha, A, Cook, R, Vogl, DT, Mangan, PA, Hummel, K, Cunningham, K, et al. Plerixafor and G-CSF versus cyclophosphamide and G-CSF for stem cell mobilization in patients with multiple myeloma. Blood (ASH Annual Meeting Abstracts). 2009 Dec; 114:Abstract 2146.Google Scholar
Shaughnessy, P, Islas-Ohlmayer, M, Murphy, J, Hougham, M, MacPherson, J, Winkler, K, et al. Plerixafor plus G-CSF compared to chemotherapy plus G-CSF for mobilization of autologous CD34+ cells resulted in similar cost but more predictable days of apheresis and less hospitalization. Blood (ASH Annual Meeting Abstracts). 2009; 114:Abstract 2277.CrossRefGoogle Scholar
Fruehauf, S, Veldwijk, MR, Seeger, T, Schubert, M, Laufs, S, Topaly, J, et al. A combination of granulocyte-colony-stimulating factor (G-CSF) and plerixafor mobilizes more primitive peripheral blood progenitor cells than G-CSF alone: results of a European phase II study. Cytotherapy. 2009; 11(8):9921001.Google Scholar
Costa, LJ, Alexander, ET, Hogan, KR, Schaub, C, Fouts, TV, Stuart, RK. Development and validation of a decision-making algorithm to guide the use of plerixafor for autologous hematopoietic stem cell mobilization. Bone Marrow Transplant. 2011; 46(1):64–9.Google Scholar
Smith, VR, Popat, U, Ciurea, S, Nieto, Y, Anderlini, P, Rondon, G, et al. Just-in-time rescue plerixafor in combination with chemotherapy and granulocyte-colony stimulating factor for peripheral blood progenitor cell mobilization. Am J Hematol. 2013; 88(9):754–7.Google Scholar
Farina, L, Spina, F, Guidetti, A, Longoni, P, Ravagnani, F, Dodero, A, et al. Peripheral blood CD34+ cell monitoring after cyclophosphamide and granulocyte-colony-stimulating factor: an algorithm for the preemptive use of plerixafor. Leuk Lymphoma. 2014; 55(2):331–6.Google Scholar
Basak, GW, Mikala, G, Koristek, Z, Jaksic, O, Basic-Kinda, S, Cegledi, A, et al. Plerixafor to rescue failing chemotherapy-based stem cell mobilization: it’s not too late. Leuk Lymphoma. 2011; 52(9):1711–9.Google Scholar
Vishnu, P, Roy, V, Paulsen, A, Zubair, AC. Efficacy and cost-benefit analysis of risk-adaptive use of plerixafor for autologous hematopoietic progenitor cell mobilization. Transfusion. 2012; 52(1):5562.Google Scholar
Li, J, Hamilton, E, Vaughn, L, Graiser, M, Renfroe, H, Lechowicz, MJ, et al. Effectiveness and cost analysis of “just-in-time” salvage plerixafor administration in autologous transplant patients with poor stem cell mobilization kinetics. Transfusion. 2011; 51(10):2175–82.Google Scholar
Cashen, AF, Lazarus, HM, Devine, SM. Mobilizing stem cells from normal donors: is it possible to improve upon G-CSF? Bone Marrow Transplant. 2007; 39(10):577–88.Google Scholar
Anderlini, P, Przepiorka, D, Seong, C, Smith, TL, Huh, YO, Lauppe, J, et al. Factors affecting mobilization of CD34+ cells in normal donors treated with filgrastim. Transfusion. 1997; 37(5):507–12.Google Scholar
Grigg, AP, Roberts, AW, Raunow, H, Houghton, S, Layton, JE, Boyd, AW, et al. Optimizing dose and scheduling of filgrastim (granulocyte colony-stimulating factor) for mobilization and collection of peripheral blood progenitor cells in normal volunteers. Blood. 1995; 86(12):4437–45.Google Scholar
Holm, M. Not all healthy donors mobilize hematopoietic progenitor cells sufficiently after G-CSF administration to allow for subsequent CD34 purification of the leukapheresis product. J Hematother. 1998; 7(2):111–3.Google Scholar
Platzbecker, U, Prange-Krex, G, Bornhauser, M, Koch, R, Soucek, S, Aikele, P, et al. Spleen enlargement in healthy donors during G-CSF mobilization of PBPCs. Transfusion. 2001; 41(2):184–9.Google Scholar
Stroncek, DF, Dittmar, K, Shawker, T, Heatherman, A, Leitman, SF. Transient spleen enlargement in peripheral blood progenitor cell donors given G-CSF. J Transl Med. 2004; 2:25.Google Scholar
Horowitz, MM, Confer, DL. Evaluation of hematopoietic stem cell donors. Hematology Am Soc Hematol Educ Program. 2005:469–75.Google Scholar
Nash, RA, Bowen, JD, McSweeney, PA, Pavletic, SZ, Maravilla, KR, Park, MS, et al. High-dose immunosuppressive therapy and autologous peripheral blood stem cell transplantation for severe multiple sclerosis. Blood. 2003; 102(7):2364–72.CrossRefGoogle ScholarPubMed
Stricker, RB, Goldberg, B. G-CSF and exacerbation of rheumatoid arthritis. Am J Med. 1996; 100(6):665–6.Google Scholar
Burt, RK, Fassas, A, Snowden, J, van Laar, JM, Kozak, T, Wulffraat, NM, et al. Collection of hematopoietic stem cells from patients with autoimmune diseases. Bone Marrow Transplant. 2001; 28(1):112.Google Scholar
Gottenberg, JE, Roux, S, Desmoulins, F, Clerc, D, Mariette, X. Granulocyte colony-stimulating factor therapy resulting in a flare of systemic lupus erythematosus: comment on the article by Yang and Hamilton. Arthritis Rheum. 2001; 44(10):2458–60.Google Scholar
Spitzer, G, Adkins, D, Mathews, M, Velasquez, W, Bowers, C, Dunphy, F, et al. Randomized comparison of G-CSF + GM-CSF vs G-CSF alone for mobilization of peripheral blood stem cells: effects on hematopoietic recovery after high-dose chemotherapy. Bone Marrow Transplant. 1997; 20(11):921–30.CrossRefGoogle ScholarPubMed
Gazitt, Y. Comparison between granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor in the mobilization of peripheral blood stem cells. Curr Opin Hematol. 2002; 9(3):190–8.Google Scholar
Rettig, MP, Shannon, WD, Ritchey, J, Holt, M, McFarland, K, Lopez, S, et al. Characterization of human CD34+ hematopoietic stem cells following administration of G-CSF or plerixafor. Blood (ASH Annual Meeting Abstracts). 2008 Dec; 112:Abstract 3476.Google Scholar
Rettig, MP, Lopez, S, McFarland, K, DiPersio, JF. Rapid and prolonged mobilization of human CD34+ hematopoietic stem cells following intravenous (IV) administration of plerixafor. Blood (ASH Annual Meeting Abstracts). 2010 Dec; 116:Abstract 2261.Google Scholar
Kymes, SM, Pusic, I, Lambert, DL, Gregory, M, Carson, KR, DiPersio, JF. Economic evaluation of Plerixafor for stem cell mobilization. Am J Manag Care. 2012; 18(1):3341.Google Scholar
Dar, A, Goichberg, P, Shinder, V, Kalinkovich, A, Kollet, O, Netzer, N, et al. Chemokine receptor CXCR4-dependent internalization and resecretion of functional chemokine SDF-1 by bone marrow endothelial and stromal cells. Nat Immunol. 2005; 6(10):1038–46.Google Scholar
Imai, K, Kobayashi, M, Wang, J, Shinobu, N, Yoshida, H, Hamada, J, et al. Selective secretion of chemoattractants for haemopoietic progenitor cells by bone marrow endothelial cells: a possible role in homing of haemopoietic progenitor cells to bone marrow. Br J Haematol. 1999; 106(4):905–11.CrossRefGoogle ScholarPubMed
Jung, Y, Wang, J, Schneider, A, Sun, YX, Koh-Paige, AJ, Osman, NI, et al. Regulation of SDF-1 (CXCL12) production by osteoblasts; a possible mechanism for stem cell homing. Bone. 2006; 38(4):497508.Google Scholar
Ponomaryov, T, Peled, A, Petit, I, Taichman, RS, Habler, L, Sandbank, J, et al. Induction of the chemokine stromal-derived factor-1 following DNA damage improves human stem cell function. J Clin Invest. 2000; 106(11):1331–9.Google Scholar
Watt, SM, Forde, SP. The central role of the chemokine receptor, CXC4R, in haemopoietic stem cell transplantation: will CXCR4 antagonists contribute to the treatment of blood disorders? Vox Sang. 2008; 94(1):1832.Google Scholar
Peled, A, Petit, I, Kollet, O, Magid, M, Ponomaryov, T, Byk, T, et al. Dependence of human stem cell engraftment and repopulation on NOD/SCID mice on CXCR4. Science. 1999; 283(5403):845–8.Google Scholar
Nagasawa, T, Hirota, S, Tachibana, K, Takakura, N, Nishikawa, S, Kitamura, Y, et al. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature. 1996; 382(6592):635–8.Google Scholar
Tachibana, K, Hirota, S, Iizasa, H, Yoshida, H, Kawabata, K, Kataoka, Y, et al. The chemokine receptor CXCR4 is essential for vascularization of the gastrointestinal tract. Nature. 1999; 393(6685):591–4.Google Scholar
Ma, Q, Jones, D, Borghesani, PR, Segal, RA, Nagasawa, T, Kishimoto, T, et al. Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4-and SDF-1-deficient mice. Proc Natl Acad Sci USA. 1998; 95(16):9448–53.Google Scholar
Ma, Q, Jones, D, Springer, TA. The chemokine receptor CXCR4 is required for the retention of B lineage and granulocytic precursors within the bone marrow microenvironment. Immunity. 1999; 10(4):463–71.Google Scholar
Robinson, JA, Demarco, S, Gombert, F, Moehle, K, Obrecht, D. The design, structures and therapeutic potential of protein epitope mimetics. Drug Discov Today. 2008; 13(21–22):944–51.Google Scholar
Schmitt, S, Weinhold, N, Dembowsky, K, Neben, K, Witzens-Harig, M, Braun, M, et al. First results of a phase-II study with the new CXCR4 antagonist POL6326 to mobilize hematopoietic stem cells (HSC) in multiple myeloma (MM). Blood (ASH Annual Meeting Abstracts). 2010 Dec; 116:Abstract 824.Google Scholar
Elices, MJ, Osborn, L, Takada, Y, Crouse, C, Luhowskyj, S, Hemler, ME, et al. VCAM-1 on activated endothelium interacts with the leukocyte integrin VLA-4 at a site distinct from the VLA-4/fibronection binding site. Cell. 1990; 60(4):577–84.Google Scholar
Scott, LM, Priestley, GV, Papayannopoulou, T. Deletion of alpha4 integrins from adult hematopoietic cells reveals roles in homeostasis, regeneration, and homing. Mol Cell Biol. 2003; 23(24):9349–60.Google Scholar
Priestley, GV, Ulyanova, T, Papayannopoulou, T. Sustained alterations in biodistribution of stem/progenitor cells in Tie2Cre+ alpha4(f/f) mice are hematopoietic cell autonomous. Blood. 2007; 109(1):109–11.Google Scholar
Priestley, GV, Scott, LM, Ulyanova, T, Papayannopoulou, T. Lack of alpha4 integrin expression in stem cells restricts competitive function and self-renewal activity. Blood. 2006; 107(7):2959–67.Google Scholar
Jing, D, Oelschlaegel, U, Ordemann, R, Holig, K, Ehninger, G, Reichmann, H, et al. CD49d blockade by natalizumab in patients with multiple sclerosis affects steady-state hematopoiesis and mobilizes progenitors with a distinct phenotype and function. Bone Marrow Transplant. 2010; 45(10):1489–96.CrossRefGoogle ScholarPubMed
Zohren, F, Toutzaris, D, Klarner, V, Hartung, HP, Kieseier, B, Haas, R. The monoclonal anti-VLA-4 antibody natalizumab mobilizes CD34+ hematopoietic progenitor cells in humans. Blood. 2008; 111(7):3893–5.Google Scholar
Foley, J. Recommendations for the selection, treatment, and management of patients utilizing natalizumab therapy for multiple sclerosis. Am J Manag Care. 2010; 16(6 Suppl):S178–83.Google Scholar
Ransohoff, RM. Natalizumab for multiple sclerosis. N Engl J Med. 2007; 356(25):2622–9.Google Scholar
Davenport, RJ, Munday, JR. Alpha4-integrin antagonism − an effective approach for the treatment of inflammatory diseases? Drug Discov Today. 2007; 12(13–14):569–76.CrossRefGoogle ScholarPubMed
Jackson, DY. Alpha 4 integrin antagonists. Curr Pharm Des. 2002; 8(14):1229–53.Google Scholar
Yang, GX, Hagmann, WK. VLA-4 antagonists: potent inhibitors of lymphocyte migration. Med Res Rev. 2003; 23(3):369–92.Google Scholar
Ghosh, S, Panaccione, R. Anti-adhesion molecule therapy for inflammatory bowel disease. Therap Adv Gastroenterol. 2010; 3(4):239–58.CrossRefGoogle ScholarPubMed
Takazoe, M, Watanabe, M, Kawaguchi, T, Matsumoto, T, Oshitani, N, Hiwatashi, N, et al. Oral alpha-4 integrin inhibitor (AJM300) in patients with active Crohn’s disease – a randomized, double-blind, placebo-controlled trial. Gastroenterology. 2009; 136(5 Suppl 1):A-181Google Scholar
Muro, F, Iimura, S, Sugimoto, Y, Yoneda, Y, Chiba, J, Watanabe, T, et al. Discovery of trans-4-[1-[[2,5-Dichloro-4-(1-methyl-3-indolylcarboxamido)phenyl]acetyl]-(4S)-methoxy-(2S)-pyrrolidinylmethoxy]cyclohexanecarboxylic acid: an orally active, selective very late antigen-4 antagonist. J Med Chem. 2009; 52(24):7974–92.Google Scholar
Ramirez, P, Rettig, MP, Uy, GL, Deych, E, Holt, MS, Ritchey, JK, et al. BIO5192, a small molecule inhibitor of VLA-4, mobilizes hematopoietic stem and progenitor cells. Blood. 2009; 114(7):1340–3.Google Scholar
King, AG, Johanson, K, Frey, CL, DeMarsh, PL, White, JR, McDevitt, P, et al. Identification of unique truncated KC/GROβ chemokines with potent hematopoietic and anti-infective activities. J Immunol. 2000; 164(7):3774–82.Google Scholar
King, AG, Horowitz, D, Dillon, SB, Levin, R, Farese, AM, MacVittie, TJ, et al. Rapid mobilization of murine hematopoietic stem cells with enhanced engraftment properties and evaluation of hematopoietic progenitor cell mobilization in rhesus monkeys by a single injection of SB-251353, a specific truncated form of the human CXC chemokine GROβ. Blood. 2001; 97(6):1534–42.CrossRefGoogle ScholarPubMed
Pelus, LM, Bian, H, King, AG, Fukuda, S. Neutrophil-derived MMP-9 mediates synergistic mobilization of hematopoietic stem and progenitor cells by the combination of G-CSF and the chemokines GROβ/CXCL2 and GROβT/CXCL2Δ4. Blood. 2004; 103(1):110–9.Google Scholar
Pelus, LM, Fukuda, S. Peripheral blood stem cell mobilization: the CXCR2 ligand GROβ rapidly mobilizes hematopoietic stem cells with enhanced engraftment properties. Exp Hematol. 2006; 34(8):1010–20.Google Scholar
Osawa, M, Hanada, K, Hamada, H, Nakauchi, H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science. 1996; 273(5272):242–5.Google Scholar
Matsuzaki, Y, Kinjo, K, Mulligan, RC, Okano, H. Unexpectedly efficient homing capacity of purified murine hematopoietic stem cells. Immunity. 2004; 20(1):8793.Google Scholar
Fukuda, S, Bian, H, King, AG, Pelus, LM. The chemokine GROβ mobilizes early hematopoietic stem cells characterized by enhanced homing and engraftment. Blood. 2007; 110(3):860–9.Google Scholar
Sackstein, R. The lymphocyte homing receptors: gatekeepers of the multistep paradigm. Curr Opin Hematol. 2005; 12(6):444–50.Google Scholar
Wodnar-Filipowicz, A. Flt3 ligand: role in control of hematopoietic and immune functions of the bone marrow. News Physiol Sci. 2003; 18:247–51.Google Scholar
He, S, Chu, J, Vasu, S, Deng, Y, Yuan, S, Zhang, J, et al. FLT3L and Plerixafor combination increases hematopoietic stem cell mobilization and leads to improved transplantation outcome. Biol Blood Marrow Transplant. 2014; 20(3):309–13.Google Scholar
Olson, JA, Leveson-Gower, DB, Gill, S, Baker, J, Beilhack, A, Negrin, RS. NK cells mediate reduction of GVHD by inhibiting activated, alloreactive T cells while retaining GVT effects. Blood. 2010; 115(21):4293–301.Google Scholar
Rezvani, K, Mielke, S, Ahmadzadeh, M, Kilical, Y, Savani, BN, Zeilah, J, et al. High donor FOXP3-positive regulatory T-cell (Treg) content is associated with a low risk of GVHD following HLA-matched allogeneic SCT. Blood. 2006; 108(4):1291–7.Google Scholar
Anandasabapathy, N, Hurley, A, Breton, G, Caskey, M, Trumpfheller, C, Sarma, P, et al. A phase 1 trial of the hematopoietic growth factor CDX-301 (rhuFlt3L) in healthy volunteers. Biol Blood Marrow Transplant. 2013; 19(2 Suppl):S112S113.Google Scholar
Hill, JM, Zalos, G, Halcox, JP, Schenke, WH, Waclawiw, MA, Quyyumi, AA, et al. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med. 2003; 348(7):593600.Google Scholar
Khan, SS, Solomon, MA, McCoy, JP Jr. Detection of circulating endothelial cells and endothelial progenitor cells by flow cytometry. Cytometry B Clin Cytom. 2005; 64(1):18.Google Scholar
Wojakowski, W, Landmesser, U, Bachowski, R, Jadczyk, T, Tendera, M. Mobilization of stem and progenitor cells in cardiovascular diseases. Leukemia. 2012; 26(1):2333.Google Scholar
Asahara, T, Takahashi, T, Masuda, H, Kalka, C, Chen, D, Iwaguro, H, et al. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J. 1999; 18(14):3964–72.Google Scholar
Schroder, K, Kohnen, A, Aicher, A, Liehn, EA, Buchse, T, Stein, S, et al. NADPH oxidase Nox2 is required for hypoxia-induced mobilization of endothelial progenitor cells. Circ Res. 2009; 105(6):537–44.Google Scholar
Lundby, C, Gassmann, M, Pilegaard, H. Regular endurance training reduces the exercise induced HIF-1α and HIF-2α mRNA expression in human skeletal muscle in normoxic conditions. Eur J Appl Physiol. 2006; 96(4):363–9.Google Scholar
Cubbon, RM, Murgatroyd, SR, Ferguson, C, Bowen, TS, Rakobowchuk, M, Baliga, V, et al. Human exercise-induced circulating progenitor cell mobilization is nitric oxide-dependent and is blunted in South Asian men. Arterioscler Thromb Vasc Biol. 2010; 30(4):878–84.Google Scholar
Hambrecht, R, Adams, V, Erbs, S, Linke, A, Krankel, N, Shu, Y, et al. Regular physical activity improves endothelial function in patients with coronary artery disease by increasing phosphorylation of endothelial nitric oxide synthase. Circulation. 2003; 107(25):3152–8.Google Scholar
Bonsignore, MR, Morici, G, Riccioni, R, Huertas, A, Petrucci, E, Veca, M, et al. Hemopoietic and angiogenetic progenitors in healthy athletes: different responses to endurance and maximal exercise. J Appl Physiol (1985). 2010; 109(1):60–7.Google Scholar
Jenkins, NT, Witkowski, S, Spangenburg, EE, Hagberg, JM. Effects of acute and chronic endurance exercise on intracellular nitric oxide in putative endothelial progenitor cells: role of NADPH oxidase. Am J Physiol Heart Circ Physiol. 2009; 297(5):H1798–805.Google Scholar

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
×