stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application:...

18
ELSEVIER Critical Reviews in Oncology/Hematology 22 (1996) 61-78 Critical Reviews in ONCOLOGY/ HEMA TOLOG Y Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application Li Lu*~*~, Rong-Nian Shenavcyd, Hal E. BroxmeyeraTbTd ‘Department of Medicine (Hematology/Oncology), Indiano University School of Medicine, Indianapolis, IN 46202-5321. USA bDepartment of Microbiology/Immunology, Indiano University School of Medicine, Indianapolis, IN 46202-5121, USA ‘Department of Radbtion Oncology, Indiano University School of Medicine, Indianapolis, IN 46202-5121, USA dThe Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202-5121. USA Accepted 28 October 1994 Contents I. 2. 3. 4. 5. 6. 7. 8. Introduction .......................................................................... Hematopoietic stem cell purification and ex vivo expansion .................................. 2.1. Stem cell purification ............................................................. 2.2. Long-term culture and ex vivo expansion ........................................... Bone marrow stem cell transplantation .................................................... Umbilical cord blood stem cell transplantation ............................................. 4. I. Quantity and quality of immature hematopoietic stem cells from umbilical cord blood .... 4.2. Immune cells in cord blood ....................................................... 4.2.1. Number and phenotype of cord blood T-lymphocytes ......................... 4.2.2. Function of the T-lymphocytes in cord blood ................................ 4.2.3. Maternal cell contamination in cord blood ................................... Peripheral blood stem cell transplantation ................................................. 5.1. Blood stem cell mobilization and collection .......................................... 5.2. Kinetics of hemopoietic reconstitution after ABSCT .................................. 5.3. Contamination of harvested blood tumor cells ..................................... 5.4. Stability and sustainment of short- and long-term hemopoietic reconstitution after ABSC T Fetal liver stem cells (FLSC) transplantation ............................................ In utero hematopoietic stem cell transplants for inherited diseases .......................... Stem cell as target for gene therapy .................................................... 8.1. Increase transduction efficiency .................................................. 8.2. Long-term gene expression in hematopoietic cells in vivo ............................ 9. Conclusion ............................................................................ Acknowledgements .............................................................................. Reviewer ....................................................................................... References ..................................................................................... Biographies ..................................................................................... 62 62 62 63 64 65 65 65 66 66 66 66 67 68 68 68 69 69 69 70 70 71 71 71 71 78 Abbreviations: ABMT, autologous bone marrow transplantation; ABSCT, autologous blood stem cell transplantation; ADA, adenosine deaminase; ALL, acute. lymphocytic leukemia; allo, allogeneic; AML, acute myeloid leukemia; BM, bone marrow; BMT, bone marrow transplantation; CB, cord blood, CBSC, cord blood stem cells; CBSCT, cord blood stem cell transplantation; CML, chronic myeloid leukemia; CSF, colony stimulating factors; CT, cytotoxic therapy; FL, fetal liver; FLSC, fetal liver stem cells; GM-CSF, granulocyte macrophage-colony stimulating factor; GVHD, graft-versus-host disease; HGF, hematopoietic growth factors; HLA, human leukocyte antigens; HPP-CFC, high proliferative potential-colony for- ming cells; HSC, hematopoietic stem cells; IL, interleukin; LTC-IC, long-term culture-initiating cells; NK, natural killer; PB, peripheral blood; PBSC, peripheral blood stem cells; PCR, polymerase chain reaction; rhu, recombinant human; SCID, severe combined immunodeficiency disease; SLF, steel factor (also termed stem cell factor). *Corresponding author, Indiana University School of Medicine, 975 W. Walnut Street, Room 501,Indianapolis, IN 46202-5 121,USA. Tel.: (317) 274-7560; fax: (317) 274-7592. 1040-8428/96/$32.000 1996Elsevier Science Ireland Ltd. All rights reserved SSDI 1040-8428(95)0179-B

Upload: li-lu

Post on 26-Jun-2016

221 views

Category:

Documents


7 download

TRANSCRIPT

Page 1: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

ELSEVIER Critical Reviews in Oncology/Hematology 22 (1996) 61-78

Critical Reviews in

ONCOLOGY/ HEMA TOLOG Y

Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

Li Lu*~*~, Rong-Nian Shenavcyd, Hal E. BroxmeyeraTbTd ‘Department of Medicine (Hematology/Oncology), Indiano University School of Medicine, Indianapolis, IN 46202-5321. USA

bDepartment of Microbiology/Immunology, Indiano University School of Medicine, Indianapolis, IN 46202-5121, USA ‘Department of Radbtion Oncology, Indiano University School of Medicine, Indianapolis, IN 46202-5121, USA

dThe Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202-5121. USA

Accepted 28 October 1994

Contents

I. 2.

3. 4.

5.

6. 7. 8.

Introduction .......................................................................... Hematopoietic stem cell purification and ex vivo expansion .................................. 2.1. Stem cell purification ............................................................. 2.2. Long-term culture and ex vivo expansion ........................................... Bone marrow stem cell transplantation .................................................... Umbilical cord blood stem cell transplantation ............................................. 4. I. Quantity and quality of immature hematopoietic stem cells from umbilical cord blood .... 4.2. Immune cells in cord blood .......................................................

4.2.1. Number and phenotype of cord blood T-lymphocytes ......................... 4.2.2. Function of the T-lymphocytes in cord blood ................................ 4.2.3. Maternal cell contamination in cord blood ...................................

Peripheral blood stem cell transplantation ................................................. 5.1. Blood stem cell mobilization and collection .......................................... 5.2. Kinetics of hemopoietic reconstitution after ABSCT .................................. 5.3. Contamination of harvested blood tumor cells ..................................... 5.4. Stability and sustainment of short- and long-term hemopoietic reconstitution after ABSC T Fetal liver stem cells (FLSC) transplantation ............................................ In utero hematopoietic stem cell transplants for inherited diseases .......................... Stem cell as target for gene therapy .................................................... 8.1. Increase transduction efficiency .................................................. 8.2. Long-term gene expression in hematopoietic cells in vivo ............................

9. Conclusion ............................................................................ Acknowledgements .............................................................................. Reviewer ....................................................................................... References ..................................................................................... Biographies .....................................................................................

62 62 62 63 64 65 65 65 66 66 66 66 67 68 68 68 69 69 69 70 70 71 71 71 71 78

Abbreviations: ABMT, autologous bone marrow transplantation; ABSCT, autologous blood stem cell transplantation; ADA, adenosine deaminase; ALL, acute. lymphocytic leukemia; allo, allogeneic; AML, acute myeloid leukemia; BM, bone marrow; BMT, bone marrow transplantation; CB, cord blood, CBSC, cord blood stem cells; CBSCT, cord blood stem cell transplantation; CML, chronic myeloid leukemia; CSF, colony stimulating factors; CT, cytotoxic therapy; FL, fetal liver; FLSC, fetal liver stem cells; GM-CSF, granulocyte macrophage-colony stimulating factor; GVHD, graft-versus-host disease; HGF, hematopoietic growth factors; HLA, human leukocyte antigens; HPP-CFC, high proliferative potential-colony for- ming cells; HSC, hematopoietic stem cells; IL, interleukin; LTC-IC, long-term culture-initiating cells; NK, natural killer; PB, peripheral blood; PBSC, peripheral blood stem cells; PCR, polymerase chain reaction; rhu, recombinant human; SCID, severe combined immunodeficiency disease; SLF, steel factor (also termed stem cell factor).

*Corresponding author, Indiana University School of Medicine, 975 W. Walnut Street, Room 501, Indianapolis, IN 46202-5 121, USA. Tel.: (3 17) 274-7560; fax: (317) 274-7592.

1040-8428/96/$32.00 0 1996 Elsevier Science Ireland Ltd. All rights reserved SSDI 1040-8428(95)0179-B

Page 2: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

62 L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (1996) 61-78

Bone marrow transplantation (BMT) has progressed rapidly during the past two decades to that of a treat- ment of choice as a therapeutically effective modality for the treatment of selected patients with malignant disease and non-malignant hematological disorders. However, its use is limited by availability of human leukocyte anti- gens (HLA)-matched donor cells, engraftment and graft-versus-host disease (GVHD). Prevention of GVHD, improvement in the speed and quality of mar- row reconstitution, and screening of new im- munomodulating agents which improve engraftment and augment hemopoiesis are intense areas of investiga- tion. To this end there has clearly been progress in puri- tication and characterization of human stem cells from different tissue sources. Discussed in this review are: (a) stem cell purification, characterization and ex vivo ex- pansion; (b) bone marrow stem cell transplantation; (c) cord blood stem cell transplantation; (d) peripheral blood stem cell transplantation; (e) fetal liver stem cell transplantation; (f) in utero stem cell transplantation; and (g) evaluation of the capacity of stem cells to serve as targets for gene therapy.

1. Illtmduction

As early as 1949, Jacobson et al, [l] showed that shielding the spleen allowed mice to recover from lethal irradiation. Shortly thereafter Lorenz et al. [2] showed that irradiation protection could be achieved by an in- travenous infusion of syngeneic marrow. Since that time, marrow grafting has been used for hematopoietic recovery, especially after myeloablative therapy using ir- radiation and/or chemotherapy. Thomas et al. [3] demonstrated that large quantities of marrow could be safely infused intravenously, but grafting was difficult. Mathe [4] reported an enduring allogeneic marrow graft in a patient with leukemia, and Gatti et al. [5] performed marrow transplantation from a matched sibling for an infant with an immunological deficiency disease. Thus, began the ‘modem’ era of bone marrow transplantation (BMT) [6]. BMT was initially developed as a treatment for severe combined immunodeficiency disease (SCID), aplastic anemia and acute leukemia. Based on pro- gressively improved results during the past two decades, this therapy has provided a means for curative correc- tion of a large number of lethal congenital and acquired disorders of the hematopoietic and lymphoid systems. BMT has become the treatment of choice for aplastic anemia and has emerged as a highly promising approach for the treatment of high-risk forms of leukemia, par- ticularly when applied to patients in remission early in the course of their disease, and for a large variety of im- munodeticiencies, hemopathies and inborn errors of me- tabolism. Intensive chemotherapy and/or irradiation followed by BMT reconstitution is being increasingly

used in the treatment of malignancies and inherited diseases. BMT represents a viable therapeutic approach, but its use is limited by the availability of HLA-matched donor cells, engraftment problems and graft-versus-host disease (GVHD).

Hematopoietic growth factors (HGFs) were initially identified by their ability to support stem/progenitor cell growth in culture assays [7,8]. During the past decade, a number of HGFs have been purified, cloned and recombinant forms produced. This has allowed clini- cians the opportunity to stimulate blood cell production in a relatively controlled fashion to achieve a therapeutic benefit. In clinical trials it has already been shown that granulocyte-macrophage (GM)colony stimulating fac- tor (CSF) and G-CSF can accelerate marrow recovery after both allogeneic and autologous marrow graft with significant shortening of the time in the hospital [9,10]. The purification, characterization and biological func- tion of hematopoietic stem cells responsible for reconstitution of marrow and blood and for indefinite cell renewal have been a targeted goal of experimental hematologists. During the last decade, much progress has been made in the purification of stem cells [ 1 l- 161. Gene transfer for the correction of genetic diseases of the marrow is an area of intense investigation. The use of retroviral vectors has increased the efficiency of gene transfer in the laboratory [17-211. Sustained expression of genes transferred into hematopoietic stem cells has been successful [ 181. Thus, stem cells can not only be used for marrow reconstitution, but can also serve as targets for gene transfer and subsequent gene therapy.

2. Hematopoietic stem cell purification and ex vivo ex- pansion

2.1. Stem cell purijkation Hematopoietic stem cells (HSC) constitute a very

small pool of undifferentiated cells that are able to maintain their number by self-renewal and have the capability to differentiate into committed progenitor cells for all lymphoid and myeloid cell lineages. They can reconstitute the hematopoietic system of a lethally irradiated recipient [l l-151. The frequency of HSC among bone marrow (BM) cells has been variously estimated at between one per 10 000 and one per 100 000 cells. The yolk sac is the first site of hemato- poiesis during ontogeny and thus logically might be the initial source of hematopoietic stem cells. Stem cells have been identified in the fetal liver (FL), thymus and bone marrow at later stages of embryogenesis [ 141. The origin of hematopoietic cells from HSC has been demonstrated using chromosomal markers [ 161, and more recently by retroviral integration markers [ 17-231. Four tissue sources of HSC are BM, umbilical cord blood (CB), FL and adult peripheral blood (PB).

Page 3: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78 63

In mouse, hematopoietic stem cells have been purified by a variety of methods, and a population of cells (Thy- I”, Lin-, Ly6A/E+) can be obtained which are approx- imately 1000 times enriched [24-261. These cells have been used for transplantation into lethally irradiated mouse recipients, and have resulted in reconstitution of myeloid and lymphoid lineages [27]. These cells have also been used in single-cell transplantation studies [25].

In humans, there have been numerous attempts to purify or enrich hematopoietic stern/progenitor cells using density gradient centrifugation, counterflow cen- trifugal elutriation, and cell sorting based on light scat- ter and antigenic determinant properties. A marker for human stem and progenitor cells, while not specific for these cells is the cell surface antigen CD34 which is pres- ent on a small fraction (< 1%) of unfractionated cells [28-571. We have enriched a population of colony form- ing cells with 47% purity from normal human bone mar- row by fluorescence - activated cell sorting with monoclonal antibodies against CD34 and major histocompatibility class II (HLA-DR) antigens [48]. Our results have confirmed that the majority of normal human bone marrow colony forming cells express deter- minants recognized by highest densities of CD34 and lower densities of HLA-DR. We then demonstrated that recombinant human (rhu) growth cytokines can act directly on the proliferation of single hematopoietic pro- genitor cells in the absence of accessory cells and serum [Sl]. Efforts have been focused on purification of imma- ture stem cells, and these cells can be further enriched using monoclonal antibodies against CD38 [32,33], CD33 [34-351, YB5.B8 (c-kit proto-oncogene product) [36], CD45 (a leukocyte common antigen isoform) [30,31], CD71 (the transferrin receptor) [43-451, HLA- DR [39,40], rhodamine 123 [41,42], CD15 [56] and LFA-1 [46].

2.2. Long-term culture and ex vivo expansion Although there is no definitive assay yet available for

human stem cells with the ability to reconstitute the hematopoietic system in vivo, in vitro assays have iden- tified cells with some characteristics of stem cells. These include long-term culture-initiating cells (LTC-IC), high proliferative potential-colony forming cells (HPP-CFC), and colony forming unit-blast. We have enriched a pop- ulation of HPP-CFC with extensive replating capacity by isolating cells with highest density of CD34 antigens (CD34+++) from human cord blood (CB) [52]. These HPP-CFC colonies came from single CD34+++ cells, and individual HPP-CFC colonies could be replated into secondary (24 dishes. Furthermore, the extensive proliferative self-renewal potential of these cells was measured by their capacity to be replated from 2’ to 3’, and 3’ to 4’ plates with maintenance of proliferative capacity. There is no evidence yet that HPP-CFC repre-

sent long-term marrow repopulating cells, but they probably belong to a stem cell compartment based on their replating capacity in vitro [52].

The hematopoietic microenvironment is the milieu provided by non-hematopoietic cells in the bone mar- row that supports and regulates hematopoiesis. Marrow stroma is composed of tibroblasts, endothelial cells, macrophages and other cells which are responsible for the production of an extracellular matrix and hematopoietic growth factors [58]. Dexter-type long- term BM cultures, which are stromal cell-dependent long-term cultures, are thought to mimic the marrow microenvironment closely [58]. Verfaillie et al. have demonstrated that primitive progenitors can differen- tiate and also be maintained when cells are co-cultured with stromal layers, but separated from the stromal layers by an 0.4 pm microporous transwell membrane [39]. Cultivation of primitive CD34+/HLA-DR- pro- genitor cells in these cultures resulted in the recovery of significantly more LTC-IC and clonogenic cells than when cells were cultured in direct contact with the stroma in a classical Dexter-type culture system. Moreover, significantly more LTC-IC and clonogenetic cells could be recovered from stroma-noncontact cul- tures supplemented with rhu interleukin (IL)-3 than from stroma-free cultures supplemented with a com- bination of 4 cytokines. An alternative approach for the growth of early cells is to supply the cytokines required for proliferation and differentiation to a stroma-free cul- ture system. Long-term cultures can be established with CD34+ cells in a stroma-free system when defined cytokines are repeatedly added [54-57,59,60]. Cyto- kines thought to be important in the induction of differ- entiation and/or proliferation of these primitive hemato- poietic progenitors include recombinant human G-CSF, GN-CSF, IL-l, IL-3, IL-6, IL-11, and steel factor (SLF, also termed stem cell factor). Bernstein et al. [29] show- ed that the combination of IL-3, G-CSF and SLF in- creased the number of single isolated CD34+Lin- cells giving rise to colonies in vitro by lo-fold. The number of colony-forming cells derived from these primitive cells increased 20- to 40-fold in the presence of these cytokines. Our recent studies have demonstrated expan- sion of a single sorted CD34+++ cell from cord blood, and compared that with 5 000 CD34+++ cells [54]. In the presence of SLF, IL-lo and IL-3, the fold expansion of nucleated cells was greater in cultures initiated with 1 cell/well (> 5 000-fold) compared to that with 5 000 cells/well (791-fold), while the fold expansion of pro- genitor cells was greater when 5 000 cells were used to initiate suspension culture. This type of single cell assay may allow identification of early acting cytokines that are produced endogenously and which act in a feeder fashion when large numbers of cells are cultured together.

Page 4: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

64 L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78

There has recently been great interest in the ex vivo expansion of hematopoietic cells for a variety of applica- tions [61]. However ex vivo maintenance and generation of functional hematopoietic stem/progenitor cells are complex processes and are poorly understood.

We have demonstrated that incubation of cord blood cells for 7 days with SLF resulted in 7.9-, 2.2- and 2.7- fold increases in numbers of CFU-GM, BFU-E and CFU-GEMM compared to starting numbers; addition of a CSF with SLF resulted in even greater expansion of those progenitors [60]. We also found that CB plasma synergized with SLF plus PIXY321, a GM-CSF/IL-3 fu- sion protein, to promote increases in cell numbers by days 14-21 and in CFU-GEMM by day 7 [62].

As briefly outlined above, recent advances in stem cell biology have resulted in the phenotypic and functional characterization of at least subsets of human hemato- poietic stem cells, and techniques to obtain relatively pure populations of these cells and to expand these cells ex vivo. However, it is not yet clear if the earliest human stem cells with long-term marrow engrafting capacity are being expanded under these conditions, since, as we previously mentioned, there is not yet an assay for these human cells. With this cautionary note, the above tech- nology may be used to design methods for autologous transplants with tumor-free stem cell grafts using positively selected hematopoietic stem cells. Autologous transplant protocols employing positively selected human stem cells are underway in patients with solid tumors.

3. Bone marrow stem cell transplantation

Allogeneic (allo) BMT is a powerful therapeutic modality for treatment of hematologic malignancies [63]. The total number of all BMTs performed worldwide is well over 40 000, and the annual figure is still increasing [64,65]. Allo-BMT is applied to a variety of diseases [66-681, including malignant hemopathies [69], aplastic myelopathies [70], homozygous hemoglo- binopathies, such as thalassemia [71], congenital im- munodeliciencies [72], and storage diseases. Two-thirds of patients requiring BMT lack HLA-matched family donors and many others therefore look for unrelated marrow donors. Although methods have improved for typing cells and more potential donors are being recruited into registries, the time for searches for unrelated marrow donors, and the possibility that no HLA-matched will be found limit to some extent such transplants. Moreover, unrelated marrow transplants are associated with high incidence of graft failure and GVHD, presumably because of donor-recipient non- identity undetectable with currently available screening procedures.

Until tumor-specific effective new agents can be

developed, increasing dose intensity may be the best way to improve therapy for patients with hematological malignancies and solid tumors who fail conventional treatment and do not have a related or unrelated HLA- matched donor. Alkylating agents, including radiation, are likely candidates for use in dose-intensive combina- tions [73]. The dose of most antineoplastic chemothera- peutic agents that may be administered is however limited largely by the toxicity to the normal marrow. Autologous bone marrow transplantation (ABMT) in- volves reinfusion of the patient’s own marrow as protec- tion from the myeloablative effects of high-dose cytotoxic chemotherapy or chemo-radiotherapy. For many tumor types, ABMT offers higher response rates than standard approaches. For leukemias and lym- phomas, response rates of 60-80% may be achieved with the potential for cure. With solid tumors, response rates range from 30% in gliomas, 50% in melanomas and colon cancer, and to more than 60% in lung cancer, and 80% in breast cancer [74]. Prolonged disease-free survi- val is possible for patients with lymphoma, Hodgkin’s disease, leukemia and breast cancer when high-dose cytotoxic therapy (CT) is followed by ABMT. Bone marrow and/or peripheral blood can be cryopreserved before high-dose CT and utilized for stem cell rescue. Clinical trials of patients with lymphoma, leukemia, Hodgkin’s disease and breast cancer have proven the importance of timing the induction and high-dose CT, and properly sequenced therapy has resulted in pro- longed disease-free survival in these patients. ABMT has become accepted therapy in selected patients with leuke- mia [75], Hodgkin’s disease, non-Hodgkin’s lymphoma [76] and neuroblastoma [77]. More recently, ABMT has also been used with encouraging results in breast car- cinoma [78], small cell carcinoma of the lung [79], and other disorders [80].

A major concern in ABMT is the possibility that clonogenic tumor cells might contaminate the harvested bone marrow resulting in recurrence of the malignant disease on infusing these cells into the host. The frequen- cy of microscopically identifiable bone marrow con- tamination range from very high in leukemias and neuroblastoma [81] to lower in solid tumors such as breast cancer [82,83].

Recently, enriched CD34+ marrow cells have been used in combination with growth factor administration post-transplant in 21 patients with advanced breast cancer [84]. Neutrophil recovery was faster in patients receiving marrow plus G-CSF in comparison with pa- tients receiving marrow plus GM-CSF or marrow only. Positively-selected enriched CD34+ marrow cells have also been used for transplantation following high dose chemotherapy f total body irradiation in 13 patients with metastatic breast cancer and 2 patients with neuroblastoma [85]. Engraftment of neutrophils was ap-

Page 5: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78 65

parent, but slower than that seen with whole marrow. The results suggest that highly enriched CD34+ cells are capable of hematopoietic reconstitution after myelo- ablative therapy [84,85] and it is possible that this proce- dure could be used as a tumor-purging technique for autologous as well as allogeneic transplantation.

4. Umbilical cord blood stem cell transplantation

Until recently, human umbilical cord blood was a usually discarded material. Cord blood is rich in hematopoietic progenitor cells, as measured in standard clonogenic assays [60,86-891. In 1989, we reported that the neonatal blood in the placenta might contain suffi- cient hematopoietic stem cells to serve as a potential source of therapeutically transplantable cells 1861. This led to the first successful experience with cord blood stem cell transplantation (CBSCT) in a patient with Fanconi anemia [90]. This young male patient was transplanted with HLA-matched cord blood from his sister and now is healthy and cured of the hematological disorder 7 years post transplantation. Engraftment was confirmed by sex chromosomal analysis of cells, ABO- antigens in erythrocytes and restriction fragment length polymorphism study of DNA. Subsequently Wagner et al. have reported an HLA-matched sibling cord blood transplantation in a child with juvenile chronic myelogenous leukemia [91]. Engraftment was confirmed by chromosomal analysis, restriction fragment length polymorphorism and polymerase chain reaction (PCR) analysis of colony forming cells from bone marrow of the recipient after transplantation. Thus, it was conclud- ed that umbilical cord blood could be considered an ef- ficacious source of sufficient stem cells for clinical hematopoietic reconstitution. Presently, over 150 child- ren have received CBSCT for the treatment of a variety of diseases [89-951, including Fanconi anemia, leukemias such as. Ph chromosome-positive chronic myeloid leukemia (CML), AML, ALL, neuroblastoma, congenital immunodeficiency, aplastic anemia, Wiskott- Aldrich syndrome, and X-linked lymproliferative syn- drome. These results led to attempts at setting up cord blood banks [96-981. Programs for the banking of unrelated CBSCs have already begun in the United States, France, United Kingdom, The Netherlands, Ger- many and Italy. While the application of CBSCs in the treatment of patients with potentially life-threatening disorders is underway, it is unlikely that individual centers will be able to completely address questions regarding the role of CBSCs in transplantation. For this reason an international cord blood transplantation registry was started [97]. CBSC can also serve as targets for gene transfer. This will be discussed in the gene ther- apy section.

Potential advantages of using cord blood relate to the high number and quality of hematopoietic stem and progenitor cells present in the circulation at birth and to the relative immune ‘immaturity’ of the newborn im- mune cells. These two issues are discussed below.

4.1. Quantity and quality of immature hematopoietic stem cells from umbilical cord blood

Single collections of cord blood can be used to engraft the hematopoietic system of children [89-991. Whether a single collection also contains sufficient cells to suc- cessfully engraft an adult remains an open question, al- though recent studies using sibling or unrelated cord blood suggests that this is possible. In vitro studies have suggested that single collections of cord blood probably do contain sufficient cells to engraft the hematopoietic system of an adult [60,88]. Engraftment will depend not only on the number of stem progenitors, but also on the quality of these cells for proliferation and self-renewal. These qualities of stem and progenitor cells appear to be high. It has been demonstrated that committed hematopoietic progenitor cells are at least as numerous in cord blood as in normal bone marrow [ 1001. The tin- ding of more erythroid and high proliferative megakaryocyte progenitors in cord blood than in nor- mal bone marrow suggests that more primitive pro- genitor cells are present in the former, results consistent with ours [60]. We [52] have purified a population of CD34+++ cells and sorted them as a single cell/well, to demonstrate that there are 8-fold more HPP-CFC with extensive replating capacity in cord blood than in adult marrow [52]. Long-term cultures have suggested that cord blood contains more early cells than bone marrow with high proliferative potential [loll. Cord blood cells can be extensively expanded in suspension culture with different combinations of cytokines [62] and we have recently demonstrated that ex vivo expansion can also be established from single primitive cells [54]. Thus, im- mature hematopoietic stem cell and progenitor cells ap- pear to be of good quality in cord blood.

4.2. Immune cells in cord blood There has been little or no GVHD detected in child-

ren receiving HLA-matched or one antigen mismatched sibling cord blood [90-951, and results using unrelated cord blood have been similar. There is evidence that immune cells from cord blood react differently than those in adults. Monocytes/macrophage and T- and B- lymphocytes are the major immune response cells. It has been reported that a low antigen-presenting ability found in CB monocytes/macrophage [102,103] is due to deficient HLA-DR antigen expression [102] and their intrinsic immaturity [103]. Newborn B cells, although, are capable of producing amounts of IgM antibody comparable with those of adult B cells, but their capaci-

Page 6: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

66 L. Lu er 01. /Critical Reviews in Oncology/Hemarology 22 (19%) 61-78

ty to produce IgG or IgA is remarkably reduced [ 1041. Cord blood T cell functions, such as helper T cell activi- ties [IOS], expression of cell surface antigens [IO61 and receptors [107], interferonr production [IO81 and killer cell activities [IO91 are decreased compared with those of adult T cells. In contrast, some other studies with T cell mitogens demonstrate that CB T cells possess mature functions compatible with adult T cells [IOS]. Thus, the functional maturity of cord blood T cells is still not com- pletely understood.

In this context, there are two issues of interest. One is the maturity of cord blood lymphocytes, and the other one is the possibility of maternal cell contamination of collected cord blood cells.

4.2.1. Number and phenotype of cord blood T- lymphocytes. In cord blood, about 50% of low density mononuclear cells express CD3, compared to 70% in adult blood [89,113]. However, phenotypic analysis of T cell subsets (CD4, helper T cells, and CDS, suppressor T cells) revealed no significant differences between adult and cord blood. CD3positive T cells are a variable sub- population representing 44.8% of lymphocytes 11123. The majority of the CD3 cells are CD3+CD8+. Newborn T cells have lower levels of both IL-2 receptors and HLA-DR than do adult T cells. The CD4-positive T cells represent 31% of lymphocytes with a great preva- lence of the CD4+/CD45RA+ population.

4.2.2. Function of the T-lymphocytes in cord blood It is of interest that little cytotoxic T cell activity is generated by cord blood cells even after primary, sec- ondary and tertiary stimulation by allogeneic cells [ 1 IO]. Moreover, even though the proliferative response to alloantigen is high in primary response, the secondary response is relatively blunted compared to that of adult T cells, suggesting a tolerant state was induced [ 1131. The nature of cytoplasmic signal transduction was examined in cord blood T cells by stimulating them with tumor promoter and calcium ionophore [I 141. The max- imal responses observed in CB T cells were much less than those of adult T cells, whereas the Con A-induced proliferative responses of these cells showed no signili- cant differences. The decreased responses of CB T cells were due to their lower efficiency in activating the cellular events in T cell activation and proliferation phase, because cord blood T cells have significantly less ability than adult T cells to produce IL-2 and express functional IL-2R complexes, especially high-affinity IL- 2R complexes, and HLA-DR. These functional characteristics may be related to the immaturity of cord T cells.

Cord blood natural killer (NK) cells are CD3- CD56+ cells and express CD2, CD7, CDII,, CD18, CD38, CD45RA and the P75 IL-2 receptor [I IS]. Only small amounts of CD3YDl6’ cells exist in cord blood [ 1161. The majority of NK cells are in CD3CDl6+CD-

56+ and CD3’CDl6CD56+ cell populations in CB as in adult blood.

Cord blood NK cell activity is low or similar to that in adult blood [ 116,117] but lymphokine activated killer cell activity (LAK) is readily induced in cord blood by cytokines such as IL-2 or IL-12 [116,117].

4.2.3. Maternal cell contamination in cord blood. Con- tamination of cord blood with maternal cells was originally considered a theoretical concern that might lead to massive GVHD. Little or no maternal cell con- tamination had been detected [I 181 and in over 150 cord blood transplants there has been little or no GVHD [89-951. PCR analysis for amplification of 2 minisatellite sequences (33.6 and MS 51) were used to evaluate the possibility of contamination [I 191. Mater- nal cells were very rarely (2”/) present within cord blood collected at birth, since they were detected in only one of 47 cases, and this was at an even lower level in the lymphocyte cell fraction (0. 1 - 1 .O%), although more sen- sitive techniques have detected maternal cells in cord blood. It is not clear what, if anything, this low level of maternal cell contamination might mean in a clinical setting.

Thus, umbilical cord blood stem cells are an altema- tive source of transplantable cells that can be used to reconstitute hematopoiesis in children with malignant and non-malignant diseases after treatment with myefoablative doses of chemotherapy. The availability of cord blood banks [97,98,120] will allow assessment of the efficacy of using cord blood to transplant the hematopoietic system of adult recipients.

5. Peripheral blood stem cell transplantation

The presence in peripheral blood of circulating stem cells (PBSC) with potential marrow repopulating ability has been considered for a while [121]. The use of autologous PBSC for transplantation has permitted pa- tients whose bone marrow is unsuitable for collection to receive high-dose radiation and/or chemotherapy for a variety of malignancies, and is considered an attractive alternative to bone marrow transplantation in those pa- tients. Autologous PBSCT offers a number of possible advantages over autologous bone marrow transplanta- tion. These include collection of blood-derived hematopoietic progenitors without the use of general anesthesia, more rapid hematopoietic recovery after marrow-ablative therapy with the potential for reduced tumor cell contamination, and the chance of performing PBSCT in patients with residual bone marrow disease or fibrosis. A large series of high-dose therapy and autologous peripheral blood stem cell transplantation for various malignancies has been reported 11221. The present status of blood stem cell transplantation has been summarized [123]. Mobilized peripheral blood

Page 7: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

L. Lu ei al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78 61

stem cells appear to produce faster neutrophil and plate- let recovery, fewer febrile days, lower blood product re- quirement and shorter periods of hospitalization, compared with bone marrow stem cells [ 124,125]. Many groups have reported on clinical use of peripheral blood progenitor cells for transplantation of patients with ma- lignant diseases [ 1261. A number of recent reports docu- ment the success of this therapeutic approach in patients with lymphoid malignancies; -[ 1271, Hodgkin’s disease [128-1311, non-Hodgkin’s lymphoma [132-1361, multi- ple melanoma [ 137,138], leukemia [139-1411, solid tumor, such as neuroblastoma [142], multiple myeloma [143,144] and breast cancer [145], and other adtive cancer [146] in adults and children.

Since autologous blood stem cell transplantation (ABSCT), and more recently allogeneic blood stem cell transplantation, have been introduced as a treatment modality, clinical research has mostly focused on the following aspects: (a) blood stem cell mobilization and collection; (b) kinetics of hemopoietic reconstitution after transplant; (c) contamination of harvested blood mononuclear cells with clonogenic tumor cells; and (d) stability and sustainment of hemopoietic reconstitution after transplant.

5.1. Blood stem cell mobilization and collection Stem cell numbers in peripheral blood are very low

compared to those in the bone marrow. Although stem cells can be collected by apharesis, this requires the pro- cessing of large volumes of blood. Amplification of PBSC, however, facilitates collection by a limited num- ber of aphareses and has several advantages since it renders autologous stem-cell rescue feasible, even in the case of bone-marrow hypocellularity or bone-marrow metastatic invasion, and avoids general anaesthesia necessary for bone marrow harvesting. Many runs of leukapheresis are required to collect sufficient numbers of circulating stem cells for complete hematopoietic reconstitution. Several manipulators have been em- ployed to increase circulating progenitor cell numbers. Mobilization of hematopoietic progenitors from bone marrow into peripheral blood can be achieved either by chemotherapy [129,130,133,135,138,149,150], hemato- poietic growth factors [ 15 1 - 1561 or by a combination of myelosuppressive chemotherapy and growth factors [145,157-1621.

It is important to find better mobilizing techniques to provide more efficient harvesting and faster hemato- poietic recovery. A large series of reports documented that PBSC can be mobilized by chemotherapy [147-1501. It has been demonstrated that even single high dose cyclophosphamide enables the collection of high numbers of hematopoietic stem cells from the peripheral blood [ 1501. Disadvantages with chemother- apy, which appear to require high doses of chemothera- peutic agents, include the frequent occurrence of fever,

infections and complications, and occasional mortality during pancytopenia after chemotherapy priming. Recently, recombinant human (rhu) growth factors, such as granulocyte (G)- or granulocyte-macrophage (GM)colony stimulating factor (CSF), have been used as stimulators of hematopoiesis in patients with neutro- penis resulting from cytoreductive therapy. Early stud- ies demonstrated higher numbers of circulating pro- genitor cells in patients receiving G-CSF or GM-CSF to mobilize peripheral-blood progenitor cells [ 15 1 - 1621. Unseparated blood mobilized by G-CSF enhanced the engraftment of platelet in patients receiving high-dose chemotherapy (1521. Patients with non-myeloid malig- nant disorders receiving G-CSF had an increased num- ber of blood granulocyte-macrophage (CFU-GM) and erythroid (CFU-E and BFU-E) progenitor cells, and a significantly shortened period of severe throm- bocytopenia. It was noted that PBSC obtained at a sin- gle leukapherasis during hematopoietic recovery after chemotherapy and G-CSF, was sufficient to restore bone marrow function [ 1321. Rapid recovery of neutro- phils was found in all patients by the administration of G-CSF following transplantation. In other reports, it was observed that the combination of myelosuppressive chemotherapy and G-CSF mobilized more PBSC than chemotherapy alone [ 159,160]. More recently, it has been suggested that hematopoietic progenitor cells mobilized into the peripheral circulation by G-CSF can be considered equivalent to those in bone marrow in their capacity to repopulate hematopoietic tissue [ 1561. Administration of G-CSF appears to be safe with few and minor side-effects. The harvesting -of peripheral blood progenitors could be performed in a blood bank setting such that general anaesthesia would be avoided. For the recipient, a faster platelet recovery may be achieved with reductions in resultant morbidity, suppor- tive care and longer periods of hospitalization.

GM-CSF has also been used for mobilization of PBSC and has resulted in efficient collections of PBSC, and has provided rapid and sustained restoration of hematopoietic function following high-dose chemother- apy [ 1551. GM-CSF in combination with chemotherapy can mobilize more PBSC than either chemotherapy or GM-CSF alone [145,160-1621. The efficacy of GM- CSF alone or in combination with peripheral blood- derived hematopoietic progenitor cells was compared as support for patients with metastatic breast carcinoma receiving high-dose chemotherapy [ 1451. The results showed that the use of autologous blood stem cell transplantation plus GM-CSF accelerated hemato- logical recovery after high-dose chemotherapy with reduced morbidity and platelet-transfusion require- ments, compared with the use of GM-CSF alone [145]. In a recent report [ 1471 results of 33 patients undergoing PBSCT were compared to 17 concurrent patients undergoing autologous marrow transplantation. PBSC

Page 8: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

68 L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78

were mobilized with high-dose cyclophosphamide, GM- CSF or with combinations of these. An important ob- servation was that PBSCT patients had fast engraftment and a IO-day shorter length of hospital stay than those undergoing autologous marrow transplantation. Al- though the authors did not show a cost analysis, it seems reasonable to assume that PBSCT offers a cost advan- tage over ABMT.

It has been suggested that it is too early to define a panel of clinical indications for blood being the pre- ferred option over marrow for autologous transplanta- tion [163]. A randomized controlled trial is the method to address this point. The most striking advantage of autologous blood over autologous marrow seems to be the shortening of pancytopenia immediately after myeloablative therapy and transplantation. In addition to mobilization of stem and progenitor cells with G-CSF and GM-CSF, trials with IL-3 have been reported [ 1641. Other growth factors such as IL- 1, IL- 11 or steel factor, alone or in combination with G-CSF or GM-CSF may be of value in mobilizing stem cells to the blood.

Of interest is a report which examined serum levels of G-CSF, M-CSF, GM-CSF, IL-3 and IL-6 in 10 children undergoing a total of 12 PBSCT procedures in which only sustained increases in serum G-CSF levels were seen early after ABSCT [ 1651. In an another report, a se- quential coordinated pattern of hematopoietic growth factor release after myeloablative chemotherapy was seen providing indications of possible mechanisms mediating hematopoietic recovery after stem cell trans- plantation [ 1661.

The future of PBSCT likely includes allogeneic transplantation and manipulation of the graft produce to provide therapeutic benefit and improve immuno- logic recovery after marrow ablative therapy. However, it is emphasized that there are not definitive data that peripheral blood, even after mobilization techniques are used, contains long-term marrow repopulating cells.

5.2. Kinetics of hemopoietic reconstitution after ABSCT Bone marrow and circulating stem cell pools are in

dynamic equilibrium with 98% of all committed pro- genitor cells located in the marrow, but only 0.1% circu- lating at any given time [167]. There is no doubt that circulating blood cells can reconstitute short-term hematopoietic function after myeloablative therapy. Whether circulating blood cells can sustain hemopoiesis lifelong in recipients of truly myeloablative therapy has not yet been determined. Correlation of determined numbers of CFU-GM infused and granulocyte recovery has been used to predict hematopoietic recovery kinetics [ 156,160]. It was found that progenitors’ mobilization with G-CSF reduced the time to granulocyte recovery by 2-28 days, and that the number of mobilized CFU- GM infused into patients predicts the recovery of granu- locyte and platelet production after ABSCT [ 1601. In

more recent studies, CD34+ cell content has been used to quantitate stem cell autografts. Correlations between CD34+ cells, CFU-GM and/or bone marrow recovery are imperfect. The CD34+ population of cells is heterogenous and many of these cells are not stem cells. CD34+ hematopoietic progenitor cells are present in the circulation at about l/10 that of the marrow and subpopulations of these cells differ in peripheral blood compared with bone marrow [168]. Immunophenotyp- ing studies have demonstrated a small portion of mobilized CD34+CD38- cells in the peripheral blood which suggests the possibility that circulating cells with long-term repopulating ability are present [ 164,169]. The CD34+38- population is immature [ 1701.

Although numbers of blood cells needed for suc- cessful short- and long-term engraftment are not yet known some have recommended 6 x lO’/mobilized mononuclear cells/kg b.w. are needed [128], whereas others suggest 8 x lo6 CD34+ blood cells/kg b.w. will produce rapid and sustained engraftment [171].

5.3. Contamination of harvested blood tumor cells It has been hypothesized that blood stem cells might

be less contaminated by residual clonogeneic tumor cells than bone marrow and therefore the risk of relapse after blood stem cell transplantation might be less [128,163]. There is however still no definite evidence to prove this hypothesis. In many cancers, malignant cells are detected in the blood, even in early disease, using sensi- tive molecular biological techniques such as cytogene- tics, PCR, FISH and long-term tumor cell cultures [ 1721. Most data are based on steady state conditions which may not be applicable during blood stem cell harvesting due to the multiple apheresis procedures used. In a recent report, it was shown that in patients with stage IV or high-risk stage II/III breast cancer, small cell or non-small cell lung cancer, and other ad- vanced malignancies, an even higher number of circula- ting tumor cells was detected after chemotherapy followed by G-CSF [ 1731. Thus caution is needed in the use of mobilized peripheral blood from patients with tumors and the question of tumor-cell purging, even of peripheral blood cells, has to be considered [174].

5.4. Stability and sustainment of short- and long-term he- mopoietic reconstitution after ABSCT

There are two issues of concern in hematopoietic re- covery after ABSCT. One is immediate recovery and the other is long-term engraftment. The most conventional way to monitor the immediate recovery is granulocyte recovery. It is obvious that granulocyte recovery is more rapid with blood versus bone marrow autografts, and this is detected when blood cells are collected after mo- bilization with chemotherapy or hematopoietic growth factors. Whether blood stem cell transplants result in long-term engraftment remains unclear. The major

Page 9: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78 69

limitation of studying this question in humans is the lack of an assay for stem cells responsible for long-term engraftment. Gene marking will probably be a way to follow engraftment and to study the correlation between engraftment and bone marrow recovery [ 175).

6. Fetal liver stem cells (FLSC) transplantation

The human fetus is immunologically immature until the sixteenth week of gestation [176]. This makes the very early fetus a potentially ideal transplant donor or recipient. The use of fetal tissue for transplantation has a number of theoretical advantages. The most important one is the potential immunologic ‘immaturity’ of fetal tissue. This means that the recognition of ‘self’ occurs during an early stage of fetal life and that tissues used before that could be immunologically privileged. During human embryogenesis, hematopoiesis is first detected in the yolk sac, then from the fifth week it is apparent in the fetal liver [ 1771, Transplants of hematopoietic stem cells derived from fetal liver could potentially recon- stitute bone marrow function in various settings of bone marrow failure including various diseases and after cancer chemotherapy. In all species studied, including mice, rats, pigs, sheep, horses and monkeys, fetal liver transplants are associated with less GVHD than transplants of bone marrow derived stem cells. Experi- mental data indicates that T- and B-lymphocytes from second trimester fetal liver have reduced immune reac- tivity compared to mature lymphocytes. It has been found that fetal liver transplants can reconstitute long- term hematopoiesis [ 1781. Some success of fetal liver transplants in humans has been confined to children with immune deficiency disorders [194-1961. Trials in aplastic anemia and leukemia were unsuccessful but were performed without sufficiently intensive immune suppression. Therefore, the fetal liver may be used for transplantation to treat hematological disorders, im- munodeficiencies or lysosomal storage disease [ 179- 1811.

Over 230 fetal tissue transplants (especially fetal liver transplants associated with syngeneic fetal thymus) have been performed to treat 62 patients with severe immuno- deficiency diseases, severe aplastic anemia or inborn er- rors of metabolism [ 1821. It has been reported that in a patient suffering from a severe combined immunodeti- ciency disease, full immunological reconstitution was obtained after fetal liver and thymus transplantation [ 180,182]. HLA typing revealed that the T cells of the patient were of donor origin, while B cells and mono- cytes were of host origin. Immunoglobulin allotype determination showed that antibodies were synthesized by host B cells indicating that transplanted T cells and recipient cells cooperated despite an HLA mismatch.

There are however serious concerns with fetal liver [ 1831. Whether one can use partial mismatched fetal

liver in a non-immunodeliciency disease is not at all clear. Moreover, it is not clear yet if one can adequately freeze and store fetal liver stem cells. Additionally, there is the constant ethical conflict that occurs when fetal liver transplants are considered.

7. In utero bematopoietic stem cell transplants for in- herited dkases

Recent advances in prenatal diagnosis now allow the diagnosis of a number of congenital hematopoietic and metabolic disorders during the first trimester of preg- nancy [184]. The immunocompetence of the early gesta- tion fetus can be compared to the child with SCID, who can often engraft with allogeneic bone marrow without the need for immunosuppressive conditioning. The goal of in utero hematopoietic stem cell (HSC) transplanta- tion is to achieve successful engraftment using a ready source of HSC with minimal conditioning. This would result in minimal short- and long-term side effects to the recipient, increased availability of donors for children with disease that might benefit from BMT, and signifi- cantly decreased cost of medical care. The use of the pre- immune fetus of animals as the recipient has permitted transplantation across immunologic barriers and xeno- grafts have now been performed [ 184-1901. In utero studies have been done on monkeys [ 191,192] and sheep [193-1971. Four potential sources of HSC for in utero transplants include bone marrow, fetal liver, mobilized peripheral blood, and cord blood. An attempt at treat- ment of a 17-week gestation human fetus with Rh in- compatibility by transplantation of T cell depleted adult marrow has been reported [ 1981. The fetus survived but there was no documentation of chimerism or engraft- ment. In contrast, using fetal donor cells, human fetuses with bare lymphocyte syndrome, SCID and thalassemia have been treated [ 199-2021.

In utero human hematopoietic stem cell transplanta- tion is now technically feasible. Three potential clinical applications of in utero transplantation are: (a) reconstitution of congenitally diagnosed hematopoietic disease; (b) gene therapy for enzyme or factor deficiency states; and (c) prenatal-specific tolerance induction for postnatal allogeneic and xenogeneic organ transplan- tation.

8. Stem cell as target for gene therapy

Retroviral mediated gene transfer has been shown by a number of investigators to be an efficient method for introducing genetic sequences into mammalian cells [203,204] and gene replacement therapy has been sug- gested for many single gene disorders [205-2071. Adeno-associated viruses (AAV) can also be used as a vehicle for gene transfer. In this article, we focus on retroviral gene transduction. Target cells for gene trans-

Page 10: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

70 L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78

fer should be able to have self-renewal capacity and dif- ferentiate into progeny cells. Hepatocytes, endothelial cells, muscle cells, keratinocytes, tibroblasts and hema- topoietic cells, including stem/progenitor cells and lym- phocytes, have been suggested as target cells. Among the most appealing somatic cells as potential gene transfer vehicles are hematopoietic stem/progenitor cells, be- cause of their wide distribution and their well- characterized capacities for proliferation, differentiation and self-renewal. In vivo and in vitro experiments in human and animals have shown that the repopulating bone marrow stem cells are an ideal target for gene transfer [208,209]. Diseases in which gene therapy may be beneficial include Fanconi anemia [90], @-thalas- semia [210], adenosine deaminase (ADA) deficiency, purine nucleoside phosphorylase deficiency [211], chronic granulomatous disease [212], leukocyte adhe- sion deficiency [213], Gaucher’s disease [214], Niemann- Pick type B [215] and metachromatic leukodystrophy [216] etc.

Children with ADA deficiency have recurrent infec- tions as well as failure to thrive because of chronic diar- rhoea and malabsorption and a high incidence of lymphoma. BMT can be curative, but most of these in- dividuals do not have a histocompatible bone marrow donor and die early in childhood. The first gene therapy was done in a Cyear-old girl with ADA deficiency in September 1990 [217]. At that time, lymphocytes isola- ted from the peripheral blood of the patient were used as target cells and a retroviral vector containing Neo and ADA cDNA were used for gene transfer. The pa- tient received gene-treated lymphocytes monthly, be- cause blood lymphocytes are mature and are contin- uously replaced from bone marrow stem/progenitor cells which are not treated. Thus, an effort at using hematopoietic stem cells as targets for gene transfer is increasing. For efficient transduction of pluripotent hematopoietic stem cells (PHSC), an amphotropic retrovirus carrying the human ADA gene has been generated [218,219]. During the past decade, in vitro experiments have indicated that hemopoietic pro- genitors from mice, rhesus monkeys and man can be transduced at high efficiency. As preclinical tests, a number of groups performed autologous bone marrow transplantation in lethally irradiated mice and rhesus monkeys using retrovirus-infected hematopoietic stem cells [220-2261. Stable long-term expression of human ADA in mice in all hematopoietic lineages following full hematopoietic reconstitution with transduced bone mar- row has been demonstrated by several groups [222-2241. The levels of human ADA expression were compatible with endogenous murine ADA protein lev- els. Long-term expression of human ADA has been demonstrated in large animal models, such as rhesus monkeys [225,226]. Recently, autologous cord blood CD34+ cells transduced with human ADA gene were infused into three children with ADA deficiency [227].

Gaucher’s disease, characterized by a deficiency of glucocerebrosidase, is treated at present by either allogenic BMT or enzyme replacement. Neither treat- ment is ideal. BMT is limited to those patients with an HLA matched donor, which is more difficult to obtain, and is associated with significant morbidity and mortali- ty. Enzyme replacement is expensive and needs to be lifelong. Transduction of the hematopoietic stem cells of the patient followed by autologous transplantation (gene therapy) could be an effective alternative treat- ment [228-2311. A retroviral vector (LG) containing the Gaucher gene has been developed and used to transduce murine hematopoietic stem cells with high efficiency. The vector has been used to infect hematopoietic cells from Gaucher patients and has been able to correct the enzyme deficiency [230].

In HIV-l infection, it may be possible to apply the technology of gene therapy to deliver anti-viral agents directly to infected cells and potentially benefit the in- fected individuals [232,233].

The problem of poor transduction efficiency of primi- tive hematopoietic progenitor cells still remains with human cells. Efforts have been taken to improve trans- duction efficiency in primitive hematopoietic stem/pro- genitor cells with long-term gene expression in hemato- poietic cells in vivo.

8.1. Increase transduction efficiency An important requirement for increasing gene trans-

fer efficiency is that the target cells must be cycling. The murine ADA gene could be transduced into hemato- poietic stem/progenitor cells from human bone marrow and umbilical cord blood and with stable expression in long-term cultures [234]. The majority of the primitive hematopoietic stem cells from bone marrow, cord blood and peripheral blood are quiescent [234,235] and are thus difficult targets for gene transfer. In the absence of growth factor preincubation of cells for prestimulation, gene transfer efficiency was only 3-10% [236-2381. Recently, many reports, including our own, have presented encouraging results on increased gene transfer efficiency by use of recombinant growth cytokines [234,235,239-2421. Enriched CD34+ cells have been used for marker gene transfer studies with high effi- ciency. Our recent results have shown that a neo gene could be transduced into a primitive population of CD34+++ cells from CB post prestimulation with Epo, steel factor (SLF), IL-3, GM-CSF and G-CSF with 49% transduction efficiency compared to 25% presorted cells [235]. The transduction efficiency was increased up to 80% when transduction was performed at the single cell level. Stable integration was evident even after four replatings of HPP-CFC colonies [241].

8.2. Long-term gene expression in hematopoietic cells in vivo

A requirement for successful gene therapy is long-

Page 11: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

L. Lu et al. /Critical Reviews in OncologylHematology 22 (19%) 61-78 11

term expression of transferred genes. Since there is no

suitable human in vivo assay available for long-term gene expression, animal models have been used for this purpose. Long-term and stable expression of introduced genes in cells derived from transduced murine bone mar- row has now been well established in several laborato- ries. The success rate for gene expression of some introduced gene sequences at 6 months reaches lOO?/ [243]. Stable long-term expression of human ADA in mice in all lineages following complete hematopoietic reconstitution with transduced bone marrow has also been demonstrated [222-2241. More recently, long-term expression has been further confirmed using larger ani- mal model of rhesus monkey [225,226].

Progress in the use of retroviral vectors as well as other newer vector systems, to transduce hematopoietic stem cells will lead to future clinical trials in patients with genetic and oncological diseases.

9. conclusloo

During the past two decades, BMT has been bene- ficial and curative for patients with many hematologic malignancies and inherited diseases and some im- munodeficiencies. The major problems that limit the success of BMT consist of recurrence of the malignancy, GVHD, infection and the paucity of matched donors for most patients. Recent interest has been focused on the transplant of stem cells from umbilical cord blood and mobilized peripheral blood. It has been suggested that the quantity and the quality of hematopoietic stem cells in cord blood are as good as, or even possibly better than, that in normal adult marrow. To establish a cryopreserved human CB bank of unrelated donors for clinical transplantation is important. The clinical obser- vations of possible lowered GVHD in the context of HLA-matched sibling cord blood suggest the possibility of crossing certain major HLA barriers. Stem cell transplantation has progressed from a highly experi- mental procedure to being accepted as a preferred form of treatment for a wide variety of disease. Research has resulted in a steady improvement in overcoming some of the problems listed above with a consequent improve- ment in long-term survival. The efficacy and applicabili- ty of transplant will increase when all of the major problems are resolved. Molecular biology and recombi- nant technology have made gene therapy possible. Stem cells will most likely be the desired targets for gene transfer and gene therapy. Although still in its infancy, human gene therapy holds considerable potential for the long-term treatment of recessive diseases, such as gene- tic inherited diseases, cancer and some immunodefi- ciencies.

Acknowledgements

Some of our studies reported in this review were sup-

ported by US Public Health Service grants R37 CA36464, ROl CA HL46549 and ROl HL 49202 to Hal E. Broxmeyer, and grants from the Phi Beta Psi Sorority to Li Lu and Rong-Nian Shen.

Reviewer

This manuscript was reviewed by Li. T. Chen, Ph.D. Department of Anatomy, University of South Florida, College of Medicine, 12901 Bruce B. Downs Blvd., MDCO06 Tampa, Florida 336124799, USA.

References

[I] Jacobson LO, Marks EK, Robson MJ, Gaston ES, Zirkle RE. The effect of spleen protection on mortality following x- irradiation. J Lab Clin Med 34:1538-1543, 1949.

[2] Lorenx E, Uphoff D, Reid TR, Shelton E. Modification of irra- diation injury in mice and guinea pigs by bone marrow injec- tions. J Nat1 Cancer Inst 12:197-201, 1951.

[S] Thomas ED, Lochte HL, Lu WC, Ferrebee JW. Intravenous in- fusion of bone marrow in patients receiving radiation and chemotherapy. N Engl J Med 257:491-496, 1957.

[4] Mathe G. Le syndrome secondarie, Pierre d’achoppement du traitement des leucemies par I ‘irradiation total suivie de trans- fusion de cellules hematopoietiques allogeniques. Diagnostic et traitement des radiolesions aigues. Geneva: O.M.S., 1964; 197-230.

[S] Gatti RA, Meuwissen HJ, Allen HD, Hong R, Good RA. Im- munological reconstitution of sex-linked lymphogenic immuno- logical deficiency. Lancet 2: 1366- 1369, 1968.

[6] Thomas ED, Storb R, Clift RA et al. Bone marrow transplanta- tion (first of two parts). N Engl J Med 292:832-843, 1975.

[7] Metcalf D. The molecular biology and functions of granulocyte-

1:

1

1

macrophage colony-stimulating- factors. Blood 67:257-267, 1986.

[8] Broxmeyer HE. The interacting effects of cytokines on hematopoietic stem and progenitor cells. In: Mertelsmann R, Herrmann F, eds. Hematopoietic Growth Factors in Clinical Application. New York: Marcel Dekker, 1990, 3-24.

[9] Brandt SJ, Peters WP, Atwater SK et al. Effect of recombinant human granulocyte-macrophage colony-stimulating factor on hematopoietic reconstitution after high-dose chemotherapy and autologous bone marrow transplantation. N Engl J Med 318:869-876, 1988.

IO] Nemunaitis J, Singer JW, Buckner CD et al. Use of recombinant human granulocyte-macrophage colony-stimulating factor in graft failure after bone marrow transplantation. Blood 76245-253, 1990.

II] Spangrude GJ, Heimfeld S, Weissman IL. Purification and characterization of mouse hematopoietic stem cells. Science 241:58-62, 1988.

121 Sutherland HJ, Eaves CJ, Eaves AC, Dragowska W, Lansdorp PM. Characterization and partial purification of human mar- row cells capable of initiating long-term hematopoiesis in vitro. Blood 741563-1570, 1989.

[13] Till JE, McCullogh EA, Siminovitch L. A stochastic model of stem cell proliferation based on the growth of spleen colony- forming cells. Proc Nat1 Acad Sci USA 51:29-36, 1964.

[14] Metcalf D, Moore MAS. Haemopoietic Cells. Amsterdam: North Holland, 1971; 362-447.

[I.51 Suda T, Suda J, Ogawa M. Single-cell origin of mouse hemopoi- etic colonies expressing multiple lineages in variable combina- tions. Proc Nat1 Acad Sci USA 806689-6693, 1983.

[14] Abramson S, Miller RG, Phillips RA. The identification in

Page 12: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

12 L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78

adult bone marrow of phnipotent and restricted stem cells of the myeloid and lymphoid systems. J Exp Med 1451567-1579, 1977.

[17] Miller AD, Bender MA, Harris EAS, Kaleko M, Gelinas RE. Design of retrovirus vectors for transfer and expression of the human /3-globin gene. J Viral 624337-4345, 1988.

[18] Williams DA, Orkin SH. Somatic gene therapy. Current status and future prospects. J Clin Invest 77:1053-1056, 1986.

[19] Lemischka IR, Raulet DH, Mulligan RC. Developmental po- tential and dynamic behavior of hematopoietic stem cells. Cell 45~917-927, 1986.

[20] Snodgrass R, Keller G. Clonal fluctuation within the haemato- poietic system of mice reconstituted with retrovirus-infected stem cells. EMBO J 63955-3960, 1987.

[21] Cape1 B, Hawley R, Covarrubias L, Hawley T, Mintz B. Clonal contributions of small numbers of retrovirally marked hematopoietic stem cells engrafted in unirradiated neonatal W/WV mice. Proc Natl Acad Sci USA 86:4564-4568, 1989.

[22] Keller G, St&grass R. Life span of multipotential hemato- poietic stem cells in vivo. J Exp Med 171:1407-1418, 1990.

[23] Jordan CT, Lemischka IR. Clonal and systemic analysis of long- term hematopoiesis in the mouse. Genes Dev 4220-232, 1990.

[24] Spangrude GJ, Johnson GR. Resting and activated subsets of mouse muhipotent stem cells. Proc Nat1 Acad Sci USA 87:7433-7437, 1990.

[25] Smith LG, Weisman IL, Heimfeld S. Clonal analysis of hematopoietic stem-cell differentiation in vivo. Proc Nat1 Acad Sci USA 88:2788-2792, 1991.

[26] Li CL, Johnson GR. Long-term hemopoietic repopulation by Thy-l”‘, Lin-, Ly6A/E+ cells. Exp Hematol 20:1309-1315, 1992.

[27] Spangrude GJ, Smith L, Uchida N et al. Mouse hematopoietic stem cells. Blood 78:1395-1402, 1991.

[28] Civin CI, Strauss LC, Brovall C, Fackler MJ, Schwartz SJ, Shaper JS. Antigen analysis of hematopoiesis. III. A hematopoietic progenitor cell surface antigen defined by a monoclonal antibody raised against KG-la cells. J Immunol 133:157-165, 1984.

1291 Bernstein ID, Andrew RG, Zsebo KM. Recombinant human stem cell factor enhances the formation of colonies by CD34+ and CD34+Lin- cells, and the generation of colony forming cell progeny from CD34+Lin- cells cultured with interleukin-3, granulocyte colony-stimulating factor, or granulocyte- macrophage colony stimulating factor. Blood 77:23 16-2321, 1991.

[30] Lansdorp PM, Sutherland HJ, Eaves CJ. Selective expression of CD45 isoforms on functional subpopulations of CD34 positive hemopoietic cells from human bone marrow. J Exp Med 172:363-366, 1990.

[31] Wognum AW, Krystal G, Eaves CT, Eaves AC, Lansdorp PM. Increased erythropoietin-receptor expression on CD3Cpositive bone marrow cells from patients with chronic myeloid leukemia. Blood 79~642~649, 1992.

[32] Issaad C, Croisille L, Katz A, Vainchenker W, Coulombel L. A murine stromal cell line allows proliferation of very primitive human CD34++/CD38- progenitor cells in long-term cultures and semi-solid assays. Blood 81:2916-2924, 1993.

1331 Terstappen LWMM, Huang S, Safford M, Landsdorp PM, Loken MR. Sequential generations of hematopoietic colonies derived from single non-lineage-committed CD34+CD38- pro- genitor cells. Blood 77:1218-1227, 1991.

[34] Andrews RG, Singer JW, Bernstein ID. Precursors of colony- forming cells in humans can be. distinguished from colony- forming cells by expression of the CD33 and CD34 antigens and light scatter properties. J Exp Med 169:1721-1731, 1989.

[35] Ema H, Suda T, Miura Y, Nakauchi H. Colony formation of

clone-sorted human hematopoietic progenitors. Blood 75: 1941-1946, 1990.

[36] Ashman LK, Cambareni AC, To LB, Levinsky RJ, Juttner CA. Expression of the YBS.B8 antigen (c-kit proto-oncogene prod- uct) in normal bone marrow. Blood 78:30-37, 1991.

1371 Leary AG, Ogawa M. Blast cell colony assay for umbilical cord blood and adult bone marrow progenitors. Blood 69953-956, 1987.

[38] Bernstein ID, Leary AG, Andrews RG, Ogawa M. Blast colony- forming cells and precursors of colony-forming cells detectable in long-term marrow culture express the same phenotype (CD33- CD34+). Exp Hematol 19:680-682, 1991.

[39] Verfaillie CM. Direct contact between human primitive hematopoietic progenitors and bone marrow stroma is not rc- quired for long-term in vitro hematopoiesis. Blood 79: 2821-2826, 1992.

[40] Brandt J, Baird N, Lu L, Srour EF, Hoffman R. Characterixa- tion of a human hematopoietic progenitor cell capable of form- ing blast cell containing colonies in vitro. J Clin Invest 82:1017-1027, 1988.

[41] Wolf NS, Kone A, Priestley GV, Bertelmez SH. In vivo and in vitro characterization of long-term repopulating primitive hematopoietic cells isolated by sequential Hoechst 33342- rhodamine 123 FACS selection. Exp Hematol 21:614-622, 1993.

[42] Udomasakdi C, Eaves CJ, Sutherland ID, Lansdorp PM. Separ- ation of functionally distinct subpopulations of primitive human hematopoietic cells using rhodamine 123. Exp Hematol 19:338-342, 1990.

[43] Lansdorp PM, Dragowska W. Long-term erythropoiesis from constant numbers of CD34+ cells in serum-free cultures in- itiated with highly purified progenitor cells from human bone marrow. J Exp Med 175:1501-1509, 1992.

[44] Mayani H, Dragowska W, Lansdorp PM. Cytokine-induced se- lective expansion and maturation of erythroid versus myeloid progenitors from purified cord blood precursor cells. Blood 81:3252-3258, 1993.

[45] Mayani H, Dragowska W, Lansdorp PM. Characterization of functionally distinct subpopulations of CD34+ cord blood cells in serum-free long-term cultures supplemented with hemato- poietic cytokines. Blood 822664-2672, 1993.

[46] Gunji Y, Nakamura M, Hagiwara T et al. Expression and func- tion of adhesion molecules on hemopoietic stem cells: CD34+LFA-I- cells are more primitive than CD34+LFA-l+ cells. Blood 80:429-436, 1992.

[47] Huang S, Terstappen LWMM. Formation of haematopoietic microenvironment and haematopoietic stem cells from single human bone marrow stem cells. Nature 360:745-749, 1992.

[48] Lu L, Walker D, Broxmeyer HE, Hoffman R, Hu W, Walker E. Characterization of adult human marrow hematopoietic pro- genitors highly enriched by two-color sorting with my10 and major histocompatibility (MHC) class II monoclonal anti- bodies. J Immunol 1391823-1829, 1987.

[49] Lu L, Briddell RA, Graham CD, Brandt E, Bruno E, Hoffman R. Effect of recombinant and purified human hematopoietic growth factors on enriched human megakaryocyte progenitor cells. Br J Haematol 70:149-156, 1988.

[50] Lu L, Leemhuis T, Srour EF, Yang YC. Human interleukin (IL)-9 specifically stimulates proliferation of CD34+++DR+CD 33- erythroid progenitors in normal human bone marrow in the absence of serum. Exp Hematol20:418-424, 1992.

[51] Kiao M, Leemhuis T, Broxmeyer HE, Lu L. Influence of com- binations of cytokines on proliferation of isolated single cell- sorted human bone marrow hematopoietic progenitor cells in the absence and presence of serum. Exp Hematol 20:276-279, 1992.

Page 13: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78 13

[52] Lu L, Xiao M, Shen R-N, Grigsby S, Broxmeyer HE. Enrich- ment, characterization, and responsiveness of single primitive CD34+++ human umbilical cord blood hematopoietic pro- genitor- with high proliferative and replating potential. Blood 81:4:-48, 1993.

1531 Lu L, Xiao M, G&by S, Wang WX, Wu B, Shen R-N, Brox- meyer HE. Comparative effects of suppressive cytokines on iso- lated single CD34+++ stem/progenitor cells from human bone marrow and umbilical cord blood plated with and without serum. Exp Hematol 21:1442-1446, 1993.

[54] Kiao M, Broxmeyer HE, Horie M, Grigsby S, Lu L. Extensive proliferative capacity of single isolated CD34+++ human cord blood cells in suspension culture. Blood Cells 20: 455-467, 1994.

(551 Migliaccio G, Migliaccio AR, Druxin ML, Giardina P-J, Zsebo K, Adamson JW. Long-term generation of colony-forming cells in liquid culture of CD34+ cord blood cells in the presence of recombinant human stem cell factor. Blood 79:2620-2627, 1992.

[56] Brandt J, Srour EF, Van Besien K, Briddell RA, Hoffman R. Cytokinedependent long-term culture of highly enriched precursors of hematopoietic progenitor cells from human bone marrow. J Clin Invest 86932-941, 1990.

[57] Srour EF, Brandt JE, Leemhuis T, Ballas CB, Hoffman R. Rela- tionship between cytokinedependent cell cycle progression and MHC class II antigen expression by human CD34+HLA-DR- bone marrow cells. J Immunol 148:815-820, 1992.

[58] Dexter TM, Coutinho LH, Spooncer E et al. Stromal cells in hemopoiesis. In: Bock G, March J, eds. Molecular Control of Hemopoiesis. Sussex, UK Wiley and Sons, 1990; 76-86.

1591 Srour EF, Brandt JE, Briddell RA, Grigsby S, Leemhuis T, Hoffman R. Long-term generation and expansion of human primitive hematopoietic progenitor cells in vitro. Blood 81: 661-669, 1993.

[60] Broxmeyer HE, Hangoc G, Cooper S et al. Growth characteristics and expansion of human umbilical cord blood and estimation of its potential for transplantation in adults. Prcc Natl Acad Sci USA 89:4109-4113, 1992.

[6l] Williams DA. Ex vivo expansion of hematopoietic stem and progenitor cells - Robbing Peter to pay Paul? Blood 81:3169-3172, 1993.

[62] Ruggieri L, Heimfeld S, Broxmeyer HE. Cytokine-dependent ex-vivo expansion of early subsets of CD34+ cord blood myeloid progenitors is greatly enhanced by cord blood plasma, but expansion of the more mature subsets of progenitors is favored. Blood Cells 20:436-454, 1994.

[63] Thomas ED. The role of marrow transplantation in the eradica- tion of malignant disease. Cancer 491963-1969, 1982.

[64] Bortin MM, Horowitz MM, Rimm AA. Increasing utilization of allogeneic bone marrow transplantation. Results of the l988- 1990 survey. Ann Intern Med 116505-512, 1992.

(651 Bortin MM, Horowitz MM, Gale RP et al. Changing trends in allogeneic bone marrow transplantation for leukemia in the 1980s. J Am Med Assoc 268607-612, 1992.

[66] O’Reilly RJ. Allogeneic bone marrow transplantation: current status and future. directions. Blood 62941-964, 1983.

(671 Thomas ED. Frontiers in bone marrow transplantation. Blood Cells 17:259-267, 1991.

(681 Marmont AM. New frontiers for allogeneic bone marrow transplantation. Bone Marrow Transplant I l:3-IO, 1993.

[69] Berman E, Little C, Gee T, O’Reilly R, Clarkson B. Reasons that patients with acute myelogenous leukemia do not undergo allogeneic bone marrow transplantation. N Engl J Med 326156-160, 1992.

(701 Bacigalupo A, Hows J, Gluckman HE et al. Bone marrow transplantation (BMT) versus immunosuppression for the treat- ment of severe aplastic anaemia (SAA): a report of the EBMT SAA working party. Br J Haematol 70:177-182, 1988.

[7l] Lucarelli G, Galimberti M, Polchi P et al. Bone marrow

transplantation in patients with thalassemia. N Engl J Med 322:417-421, 1990.

1721 Marmont AM. Autoimmunity and allogeneic bone marrow transplantation. Bone Marrow Transplant 9: l-3, 1992.

[73] Herxig RH. The role of autologous bone marrow transplanta- tion in the treatment of solid tumors. Semin Oncol l9:7-12, 1992.

1741 Cheson BD, Lacema L, Leyland-Jones B, Sarosy G, Wittes RE. Autologous bone marrow transplantation. Current status and future directions. Ann Intern Med I l&51-65, 1989.

[75] Reiffers J, leverger G, Castaignes et al. Autologous blood stem cell transplantation in patients with hematologic malignancies. In: European Bone Marrow Transplantation Group. Cha- monix, France: European Bone Marrow Transplantation Group, 1988; 167-168.

[76] Moormeier JA, Williams SF, Kaminer LS, Gamer M, Bitran JD. High-dose tri-alkylator chemotherapy with autologous stem cell rescue in patients with refractory malignancies. J Natl Cancer Inst 8229-34, 1990.

[77] Philip T, Bernard JL, Zucher JM et al. High dose. chemoradio- therapy with bone marrow transplantation as consolidation treatment in neuroblastoma: an unselected group of stage IV pa- tients over one year of age. J Clin Oncol 5:266-271, 1987.

[78] Peters WP, Shpall EJ, Jones RB et al. High-dose combination cyclophosphamide, cisplatin and carmustine (BCNU) with bone marrow support as initial treatment for metastatic breast cancer: three-six year follow-up. Proc ASCO 9:3lA, 1990.

[79] Ihde DC, Deisseroth AB, Lichter AS et al. Late intensive com- bined modality therapy followed by autologous bone marrow infusion in extensive stage small-cell lung cancer. J Clin Oncol 41443-1454, 1986.

(801 Wolff SN, Herzig RH, Fay JW et al. High dose N,N’,N”- triethylenethiophosphoramide (Thiotepa) with autologous bone marrow transplantation: phase I studies. Semin Oncol I7 (Suppl 3):2-6, 1990.

[8l] Moss TJ, Reynolds P, Sather HN, Romansky SG, Hammond GD, Seeger RC. Prognostic value of immunocytologic detection of bone marrow metastases in neuroblastoma. N Eng J Med 324219-226, 1991.

[82] Shpall EJ, Jones RB, Bast RC et al. 4-Hydroperoxycyclophos- phamide purging of breast cancer from the mononuclear cell fraction of bone marrow in patients receiving high-dose chemo- therapy and autologous marrow support: a phase I trial. J Clin Oncol 985-93, 1991.

[83] Vaughan WP, Mann SL, Garvey J et al. Breast cancer detected in cell culture of histologically negative bone marrow predicts systemic relapse in patients with stage I, II, III and locally recur- rent disease. Proc ASCO 9:27A, 1990.

[84] Bensinger W, Andrews R, Berenson R, Buckner CD, Berstein I, Hansen J. Engraftment kinetics after transplantation of CD34 enriched marrow cells. Bone Marrow Transplant l&3, 1992.

[SS] Berenson R, Bensinger W, Hill R et al. Engraftment after infu- sion of CD34+ marrow cells in patients with breast cancer or neuroblastoma. Blood 77:1717-1722, 1991.

[86] Broxmeyer HE, Douglas DW, Hangoc G et al. Human umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cells. Proc Natl Acad Sci USA 863828-3832, 1989.

[87] Broxmeyer HE, Gluckman E, Auerbach A et al. Human umbili- cal cord blood: a clinically useful source of transplantable hematopoietic stem/progenitor cells. Int J Cell Clon 8:76-91, 1990.

[88] Broxmeyer HE, Cooper S, Yoder M, Hangoc G. Human umbili- cal cord blood as a source of transplantable hematopoietic stem and progenitor cells. Curr Top Microbial Immunol 177:195- 204, 1992.

(891 Broxmeyer HE, Lu L, Gaddy J, Ruggieri L, Srivastava A,

Page 14: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

74 L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78

Ridon G. Human umbilical cord blood transplantation: expan- sion and gene therapy of hematopoietic stem and progenitor cells and immunology. In: Levitt DJ, ed. Hematopoietic Stem Cells, Biology and Therapeutic Applications. Marcel Dekker Inc., New York, pp. 297-317, 1994.

[90] Gluckman E, Broxmeyer HE, Auerbach A et al. Hematopoietic reconstitution in a patient with Fanconi’s anemia by means of umbilical cord blood from an HLA-identical sibling. N Engl J Med 321:1174-1178, 1989.

[91] Wagner JE, Broxmeyer HE, Byrd RL et al. Transplantation of umbilical cord blood after myeloablative therapy: analysis of engraftment. Blood 79:1874-1881, 1992.

[92] Wagner JE, Kernan NA, Steinbach M, Broxmeyer HE, Gluckman E. Atlogeneic sibling cord blood transplantation in forty-four children with malignant and non-malignant disease. Lancet 346:214-219, 1995.

[93] Bogdanic V, Nemet D, Kastelan A et al. Umbilical cord blood transplantation in a patient with Philadelphia chromosome- positive chronic myeloid leukemia. Transplant 56:477-479, 1993.

(941 Vowels MR, L-P-Tang R, Berdoukas V et al. Brief report: cor- rection of x-linked lymphoproliferative disease by transplanta- tion of cord-blood stem cells. N Engl J Med 329~1623-1625, 1993.

[95] Vilmer E, Sterkers G, Rahimy C. HLA-mismatched cord blood transplantation in a patient with advanced leukemia. Transplan- tation 53:1155-1157, 1992.

[96] Rubinstein P. Placental blood-derived hematopoietic stem ceils for unrelated bone marrow reconstitution. J Hematother 2:207-210, 1993.

(971 Gluckman E, Devergie A, Thierry D et al. Clinical applications of stem cell transfusion from cord blood and rationale for cord blood banking. Bone Marrow Transplant 9:114-l 17, 1992.

[98] Gluckman E, Wagner J, Hows J, Keman N, Bradley B, Brox- meyer HE. Cord blood banking for hematopoietic stem cell transplantation: an international cord blood transplant registry. Bone Marrow Transplant 11:199-200, 1993.

[99] Broxmeyer HE, Hangoc G, Cooper S. Clinical and biological as- pects of human umbilical cord blood as a source of transplant- able hematopoietic stem and progenitor cells. Bone Marrow Transplant 9:7-IO, 1992.

[lOO] Hows JM, Marsh JCW, Bradley BA et al. Human cord blood: a source of transplantable stem cells? Bone Marrow Transplant 9~105-108, 1992.

[loll Hows JM, Bradley BA, Marsh JCW et al. Growth of human umbilicalcord blood in long-term haemopoietic cultures. Lancet 340:73-76, 1992.

[102] Stiehm ER, Sztein MB, Steeg PS et al. Deficient DR antigen ex- pression on human cord blood monocytes: reversal with lym- phokines. Clin Immunol Immunopathol 30:430-436, 1984.

1103) Taylor S, Bryson YJ. Impaired production of y-interferon by newborn cells in vitro is due to a functionally immature macro- phage. J Immunol 134:1493-1497, 1985.

[104] Anderson U, Bird AG, B&ton S, Palacios R. Humoral and cellular immunity in human studied at the cell level from birth to two years of age. Immunol Rev 57:5-38, 1981.

[IO51 Miyawaki T, Moriya N, Nagaoki T, Taniguchi N. Maturation of B-cell differentiation ability and T-cell regulatory function in infancy and childhood. Immunol Rev 57:61-87, 1981.

[IO61 Foa R, Giubellino MC, Fierro MT, Lusso P, Ferrando ML. Im- mature T lymphocytes in human cord blood identified by mono- clonal antibodies: a model for the study of the differentiation pathway of T cells in humans. Cell Immunol89: 194-201, 1984.

[IO71 Yokoi T, Miyawaki T, Yachie A, Ohzeki S, Tanigychi N. Dis- crepancy in expression ability of Tat antigen and Ia deter- minants defined by monoclonal antibodies on activated or

cultured cord blood T lymphocytes. J Immunol 129:1441-1445, 1982.

11081 Wilson CB, Westall J, Johnston L, Lewis DB, Dower SK, Alpert AR. Decreased production of interferon-gamma by human neo- natal cells. Intrinsic and regulator deficiencies. J Clin Invest 77:860-867, 1986.

[IO91 Ueno Y, Miyawaki T, Seki H et al. Differential effects of recom- binant human interferon-r and interleukin 2 on natural killer cell activity of peripheral blood in early human development. J Immunol 135:180-184, 1985.

[110] Risdon G, Gaddy J, Broxmeyer HE. Proliferative and cytotoxic responses of human cord blood T lymphocytes following allo- geneic stimulation. Cell Immunol I54:14-24, 1994.

11 111 Hannet I, Erkeller-Yuksel F, Lydyard P, Deneys V, De.Bruyere M. Developmental and maturational changes in human blood lymphocyte subpopulations. Immunol Today 13:215-218, 1992.

[I 121 Rabian-Henog C, Lesage S, Gluckman E, Charron D. Characterization of lymphocyte subpopulations in cord blood. J Hematother 2:255-257, 1993.

[ 1131 Risdon G, Gaddy J, Horie M, Broxmeyer HE. Ailoantigen priming induces a state of unresponsiveness in human cord blood T cells. Proc NatI Acad Sci USA 922413-2417, 1995.

[I 141 Matsuzaki N, Saji F, Okada T et al. Demonstration of fimction- al immaturity of signal transduction pathways in human cord T cells. Cell Immunol 124252-263, 1989.

[ 1151 Phillips JH, Hori T, Nagler A, Bhat N, Spits H, Lanier LL. On- togeny of human natural killer (NK) cells: fetal NK cells mediate cytolytic function and expnss cytoplasmic CD3 ep- silon, delta proteins. J Exp Med 175:1055-1066, 1992.

[I 161 Gaddy J, Risdon G, Broxmeyer HE. Cord blood natural killer cells are functionally and phenotypically immature but readily respond to IL-2 and IL-12. J. Inteferon and cytokine Res 15:527-536, 1995.

[I171 Keever CA. Characterization of cord blood lymphoctye sub- populations. J Hematother 2:203-206, 1993.

[118] Broxmeyer HE, Kurtzberg J, Gluckman E et al. Umbilical cord blood hematopoietic stem and repopulating cells in human clini- cal transplantaiton. Blood Cells 17:313-329, 1991.

[I191 Socie G, Gluckman E, Carosella E, Brossard Y, Lafon C, Brison 0. Search for maternal cells in human umbilical cord blood by polymerase. chain reaction amplification of two minisatellite sequences. Blood 83:340-344, 1994.

[I201 Rubinstein P, Rosenfeld RE, Adamson JW, Stevens CE. Stored placental blood for unrelated bone marrow reconstitution. Blood 84~1679-1690, 1993.

[I211 Korbling M, Fliedner TM, Holle R et al. Autologous blood stem cell (ABSCT) versus purged bone marrow transplantation (pABMT) in standard risk AML: influence of source and cell composition of the autograft on hemopoietic reconstitution and disease-free survival. Bone Marrow Transplant 7:343-349, 1991.

[I221 Anon. Proceedings of the International Symposium of Peripheral Blood Stem Cell Autografts (15-17 October 1989, Mulhouse, France). Bone Marrow Transplant 5 (Suppl), 1990.

[123] Gale RP, Henon P, Juttner C. Blood stem cell transplants come of age. Bone Marrow Transplant 9:151-155, 1992.

[I241 Chao NJ, Schriber JR, Grimes K et al. Granulocyte colony- stimulating factor ‘mobilized’ peripheral blood progenitor cells accelerate granulocyte and platelet recovery after high-dose chemotherapy. Blood 81:2031-2035, 1993.

[125] Elias AD, Ayash L, Andrerson KC et al. Mobilization of peripheral blood progenitor cells by chemotherapy and granulocyte-macrophage colony-stimulating factor for hemato- logic support after high-dose intensification for breast cancer. Blood 79:3036-3044, 1992.

[126] Kessinpr A, Armitage JO. The evolving role of autologous

Page 15: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78 15

peripheral stem cell transplantation following high-dose therapy for malignancies. Blood 77:211-213, 1991.

[ 1271 Vose JM, Kessinger A, Kennedy BC, Armitage JO, Bierman PJ. Long-term sequelae of autologous bone marrow or peripheral stem cell transplantation for lymphoid malignancies. Cancer 69:784-789, 1992.

[128] Korbling M, Holle R, Haus R et al. Autologous blood stem cell transplantation in patients with advanced Hodgkin’s disease and prior radiation to the pelvic site. J Clin Gncol 8:978-985, 1!390.

[129] Kessinger A, Bierman PJ, Vase JM, An&age JO. High-dose cyclophosphamide, carmustine, and etoposide followed by autologous peripheral stem cell transplantation for patients with relapsed Hodgkin’s disease. Blood 77:2322-2325, 1991.

11301 Hurd DD, Bostrom B, Haade RJ et al. Treatment of refractory and relapsed Hodgkin’s disease: intensive chemotherapy and autologous bone marrow or peripheral blood stem cell support. Med Pediat Gncol 18447-453, 1990.

[I311 Haas R, Hohaus S, Goldschmidt H, Witt B, Hunstein W. Autologous blood stem cell transplantation in Hodgkin’s disease - the impact of rhG-CSF on blood stem cell collection. Bone Marrow Transplant l&S, 1992.

[I321 Pettengell R, Morgenstem GR, Woll PJ et al. Peripheral blood progenitor cell transplantation in lymphoma and leukemia using a single apheresis. Blood 82:3770-3777, 1993.

[133] Kotasek D, Shepherd KM, Sage RE et al. Factors affecting blood stem cell collections following high-dose cyclophospha- mide mobilization in lymphoma, myeloma and solid tumors. Bone Marrow Transplant 911-17, 1992.

[134] Advani R, Lambom KR, Chao N et al. Granulocyte- macrophage colony-stimulating factor (GM-CSF) as an adjunct to autologous hemopoietic stem cell transplantation for lym- phoma. Ann Inter Med 116183-189, 1992.

[135] Brice P, Adam M, Marolleau JP et al. Autologous peripheral blood stem cell transplantation after high dose therapy in pa- tients with advanced lymphomas. Bone Marrow Transplant 9337-342, 1992.

[136] Kessinger A, Bierman PJ, Vose JM, Annitage JO. High-dose therapy and autologous peripheral stem cell transplantation for patients with bone marrow metastases and relapsed lymphoma: an alternative to bone marrow purging. Exp Hematol 19:1013-1016, 1991.

[l 37 ‘1 Jagannath S, Crowley J, Vesole DH, Barlogie B, Glenn L. Low- risk intensive therapy for multiple myeloma with combined autologous bone marrow and blood stem cell support. Blood 180:1666-1672, 1992.

[ 1381 Ferman J-P, Levy Y, Gerota J et al. Treatment of aggressive multiple myeloma by high-dose chemotherapy and total body ir- radiation followed by stem cell autologous graft. Blood 73:20-23, 1989.

(I391 Brim-Babapulle F, Dowding C, Bowcock SJ et al. Autografting for patients with chronic myeloid leukaemia in chronic phase: peripheral blood stem cells may have a finite capacity for main- taming haemopoiesis. Br J Haematol 73:76-81, 1989.

[MO] Reiffers J, Cony-Makhoul P, Trouette R et al. Autologous blood stem ceil transplantation for chronic granulocytic leukaemia in transformation: a report of 47 cases. Br J Haematol 77:339-345, 1991.

[141] To LB, Dyson PG, Bradford A et al. Peripheral blood stem cells collected in very early remission produce rapid and sustained autologous hematopoietic reconstitution in acute non- lymphoblastic leukaemia. Bone Marrow Transplant 2:103-108, 1987.

[I421 Dollorso S, Rivabella L, Lanino E et al. Peripheral blood stem cell (PBSC) autografts in pediatric solid tumors. Bone Marrow Transplant l&45, 1992.

11431 Fermand JP, Levy Y, Gerota J et al. Treatment of aggressive multiple myeloma by high-dose chemotherapy and total body ir- radiation followed by blood stem cell autologous graft. Blood 73:20-23, 1989.

[144] Ventura GJ, Barlogie B, Hester JP et al. High dose cyclophos- phamide, BCNU and VP-16 with autologous blood stem cell support for refractory multiple myeloma. Bone Marrow Trans- plant 5:265-268, 1990.

11451 Kritx A, Crown JP, Motxer RJ et al. Beneficial impact of peripheral blood progenitor cells in patients with metastatic breast cancer treated with high-dose chemotherapy plus granulocyte-macrophage colony-stimulting factor. A rando- mixed trial. Cancer 71:2515-2521, 1993.

[I461 Takaue Y, Watanabe T, Abe T et al. Experience with peripheral blood stem cell collection for autografts in children with active cancer. Bone Marrow Transplant 10:241-248, 1992.

(I471 Rosenfeld CS, Gremba C, Shadduck RK, Zeigler ZR, Nemunaitis J. Engraftment with peripheral blood stem cells using noncontrolled-rate cryopreservation: comparison with autologous bone marrow transplantation. Exp Hematol 22:290-294, 1994.

[I481 Kessinger A, Arm&age JO, Landmark JD, Smith DM, Weisen- burger DD. Autologous peripheral hematopoietic stem cell transplantation restores hematopoietic function following mar- row ablative therapy. Blood 71:723-727, 1988.

[149] Williams SF, Bitran JD, Richards JM et al. Peripheral blood- derived stem cell collections for use in autologous transplanta- tion after high dose chemotherapy: an alternative approach. Bone Marrow Transplant 5:129-133, 1990.

[150] To LB, Shepperd KM, Haylock DN et al. Single high doses of cyclophosphamide enable the collection of high numbers of he- mopoietic stem cells from the peripheral blood. Exp Hematol 18442-447, 1990.

[151] Gianni AM, Tarella C, Siena S et al. Durable and complete hematopoietic reconstitution after autografting of rhuGM-CSF exposed peripheral blood progenitor cells. Bone Marrow Trans- plant 6:143-145, 1990.

[152] Sheridan WP, Begley CG, Juttner CA et al. Effect of peripheral- blood progenitor cells mobilised by tilgrastim (G-CSF) on plate- let recovery after high-dose chemotherapy. Lancet 339640-644, 1992.

(153) Duhrsen U, Villeval JL, Boyd J, Kannourakis G, Morstyn G, Metcalf D. Effects of recombinant granulocyte colony- stimulating factor on hematopoietic progenitor cells in cancer patients. Blood 72:2074-2081, 1988.

[154] Haas R, Ho AD, Bredthauer U et al. Successful autologous transplantation blood stem cells mobilized with recombinant human granulccyte macrophage colony-stimulating factor. Exp Hematol 18:94-98, 1990.

[155] Bishop MR, Anderson JR, Jackson JD et al. High-dose therapy and peripheral blood progenitor cell transplantation: effects of recombinant human granulocyte-macrophage colony-stimul- ating factor on the autograft. Blood 83:610-616, 1994.

[156] Baumann I, Testa NG, Lange C et al. Haemopoietic cells mobilised into the circulation by lenograstim as alternative to bone marrow for allogeneic transplants. Lancet 341:369, 1993.

[ I571 Kanx L, Brugger W, Mertelsmann R. Mobilization of peripheral blood progenitor cells by hemopoietic growth factors following standard dose chemotherapy. Bone Marrow Transplant 10:6, 1992.

[I581 Fukuda M, Miyajima Y, Matsuyama T. Recombinant human granulocyte colony-stimulating factor enhances the release of peripheral blood stem cells following chemotherapy in childhood malignancies. Acta Haematol Japanica 53:916-922,

[159] L%rna T, Inaba S, Harada M et al. Cytotoxic drug and cyto-

Page 16: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

76 L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78

toxic drug/G-CSF mobilization of peripheral blood stem cells and their use for autografting. Bone Marrow Transplant l&215-220, 1992.

[ 1601 Kawano Y, Hirao A, Takaue Y et al. Effects of progenitor cell dose and preleukapheresis use of human recombinant granulo- cyte colony-stimulating factor on the recovery of hematopoiesis after blood stem cell autografting in children. Exp Hematol 21:103-108, 1993.

[I611 Siena S, Bregni M, Brando B, Ravagnani F, Bonadonna G, Gianni AM. Circulation of CD34+ hematopoietic stem cells in the peripheral blood of high-dose cyclophosphamide-treated pa- tients: enhancement by intravenous recombinant human granulocyte-macrophage colony-stimulating factor. Blood 741905-1914, 1989.

] 16;; Socinski MA, Cannistra SA, Elias A, Antman KH, Schnipper L, (intim JD. Granulocyte-macrophage colony-stimulating factor expands the circulating haemopoietic progenitor cell compart- ment in man. Lancet 1:1194-l 198, 1988.

(1631 Korbling M, Juttner C, Henon P, Kessinger A. Autologous blood stem cell versus bone marrow transplantation. Bone Mar- row Transplant 10:144-148, 1992.

[164] Brugger W, Brass K, Frisch J et al. Mobilization of peripheral blood progenitor cells by sequential administration of interleukin-3 and granulocyte-macrophage colony-stimulating factor following polychemotherapy with etoposide, ifosfamide and cisplatin. Blood 79:1193-1200, 1992.

[165] Kawano Y, Takaue Y, Saito S-I et al. Granulocyte colony- stimulating factor (CSF), macrophage-CSF, granulocyte- macrophage CSF, interleukin-3 and interleukin-6 levels in sera from children undergoing blood stem cell autografts. Blood 81:856-860, 1993.

[I661 Baiocchi G, Scambia G, Benedetti P et al. Autologous stem cell transplantation: sequential production of hematopoietic cytokines underlying granulocyte recovery. Cancer Res 53:1297-1303, 1993.

[I671 Fliedner TM, Steinbach KH. Repopulating potential of hematopoietic precursor cells. Blood Cells 14393-410, 1988.

[168] Bender JG, Unvetzagt KL, Walker DE et al. Identification and comparison of CD34+ cells and their subpopulations from nor- mal peripheral blood and bone marrow using multicolor flow cytometry. Blood 77:2591-2596, 1991.

[I691 Bender JG, Williams SF, Myers S et al. Characterization of chemotherapy mobilized peripheral blood progenitor cells for use in autologous stem cell transplantation. Bone Marrow Transplant 10~281-285, 1992.

[170] Terstappen LWMM, Huang S, Safford M, Lansdorp PM, Loken MR. Sequential generations of hematopoietic colonies derived from single non-line-age-committed CD34+CD38- pro- genitor cells. Blood 77:1218-1227, 1991.

[I711 Gianni AM, Siena S, Bregni M et al. Granulocyte-macrophage colony stimulating factor to harvest circulating haemopoietic stem cells for autotransplantation. Lancet 2:580-585, 1989.

[I721 Sharp JG, Vaughn WP, Kessinger A et al. Significance of detec- tion of tumor cells in hematopoietic stem cell harvests of pa- tients with breast cancer. In: Dicke KD, Armitage JO, Dicke-Evinger MJ, eds. Autologous Bone Marrow Transplanta- tion. The University of Nebraska Medical Center Publications, 1991; 3X5-391.

[ 1731 Brugger W, Bross KJ, Glatt M, Weber F, Mertelsmann R, Kanx L. Mobilization of tumor cells and hematopoietic progenitor cells into peripheral blood of patients with solid tumors. Blood 83:636+X0, 1994.

[I741 Korbling M, Fliedner TM, Holle R et al. Autologous blood stem cell (ABSCT) versus purged bone marrow transplantation @ABMT) in standard risk AML: inBuence of source and cell composition of the autograft on hemopoietic reconstitution and disease-free survival. Bone Marrow Transplant 7:343-349, 1991.

11751 Brenner MK, Rill DR, Moen RC et al. Applications of gene marking prior to autologous bone marrow transplantation. J Cell Biochem 16A (Abstr DO29):179, 1992.

[I761 Cromblehohne TM, Langer JC, Harrison MR, Zanjani ED. Transplantation of fetal cells. Am J Obstet Gynecol 164~218-230, 1991.

[177] Gabbianelli M, Broccoli G, Cianetti L, Russo G, Testa U, Peschle C. HLA expression in hematopoietic development. Class I and II antigens are induced in the definitive erythroid lineage and differentially modulated by fetal liver cytokines. J Immunol 144:3354-3360, 1990.

(1781 Gale RP. Fetal liver transplants. Bone Marrow Transplant 9:118-120, 1992.

[179] Touraine J-L. Bone-marrow and fetal-liver transplantation in immunodeficiencies and inborn errors of metabolism: lack of significant restriction of T-cell function in long-term chimeras despite HLA-mismatch: Immunol Rev 71:103-121, 1983.

[180] Roncarolo M-G, Bacehetta R. T cell repertoire and tolerance after fetal stem cell transplantation. Bone Marrow Transplant 9:127-128, 1992.

(1811 Roncarolo MG, Touraine J-L, Banchereau J. Cooperation be- tween major histocompatibility complex mismatched mononu- clear cells from a human chimera in the production of antigen-specific antibody. J Clin Invest 77:673-680, 1986.

[ 1821 Touraine J-L, Roncarolo MG, Royo C, Touraine F. Fetal tissue transplantation, bone marrow transplantation and prospective gene therapy in severe immunodeficiencies and enzyme deticien- ties. Thymus l&75-87, 1987.

[183] O’Reily RJ, Kirkpatrick D, Kapoor N et al. A comparative re- view of the results of transplants of fully allogeneic fetal liver and HLA-haplotype. mismatched T-cell depleted marrow in the treatment of severe combined immunodeBciency. In: Gale RP, Touraine JL, Lucarelli G, eds. Fetal Liver Transplantation. New York: Alan R. Liss Inc., 1985; 327-342.

[I841 Flake AW, Harrison MR, Zanjani ED. In utero stem cell transplantation. Exp Hematol 19:1061-1064, 1991.

[I851 Flake AW, Harrison MR, Ad&k NS, Zanjani ED. Transplan- tation of fetal hematopoietic stem cells in utero: the creation of hematopoietic chimeras. Science 233:776-778, 1986.

[I861 Lubin I, Faktorowich Y, Lapidot T et al. Engraftment and de- velopment of human T and B cells in mice after bone marrow transplantation. Science 252:427-431, 1991.

(1871 McCtme JM, Namikawa R, Kameshima H, Shuhz LD, Lieber- man M, Weissman IL. The SCID mouse: murine model for the analysis of human hematolymphoid differentiation and func- tion. Science 241:1632-1639, 1988.

[I881 Mosier DE, Gulixia RJ, Baird SM, Wilson DB. Transfer of a functional human immune system to mice with severe combined immunodeficiency. Nature 335:256-259, 1988.

[189] Krams SM, Dorshkind K, Gershwin EM. Generation of Bbrillary lesions after transfer of human lymphocytes into severe combined immunodeficient (SCID) mice. J Exp Med 170:1919-1930, 1989.

[190] Kamel-Reid S, Dick JE. Engraftmment of immune-deficient mice with human hematopoietic stem cells. Science 242:1706- 1709, 1988.

[191] Duncan BW, Harrison MR, Zanjani ED et al. Immunologic evaluation of hematopoietic chimetic rhesus monkeys. Trans- plant Proc 23841-843, 1991.

[192] Harrison MR, Slotnick RN, Crombleholme TM, Globus MS, Tarantal AF, Zanjani ED. In utero transplantation of fetal liver haematopoietic stem cells in monkeys. Lancet 21425-1427, 1989.

[193] Crombleholme TM, Harrison MR, Zanjani ED. In utero transplantation of hematopoietic stem cells in sheep: the role of T cells in engraftment and graft-versus-host disease. J Pediatr Surg 25:885-892, 1990.

(1941 Zanjani ED, Pallavacini MG, Ascensao JL, Flake AW, Har-

Page 17: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78 II

rison MR, Tavassoli M. Human-ovine xenogenic transplanta- tion of stem cells in utero. Bone Marrow Transplant 9:86-89, 1992.

[I951 Zanjani ED, Ascensao JL, Harrison MR, Tavassoli M. Ex vivo incubation with growth factors enhances the engraftment of fetal hematopoietic cells transplanted in sheep fetuses. Blood 79:3045-3049, 1992.

[I961 Srour EF, Zanjani ED, Brandt JE et al. Sustained human hema- topoiesis in sheep transplanted in utero during early gestation with fractionated adult human bone marrow cells. Blood 79:1404-1412, 1992.

[I971 Srour EF, Zanjani ED, Cometta K et al. Persistence of human multilineage, self-renewing lymphohematopoietic stem cells in chime& sheep. Blood 82:3333-3342, 1993.

[I981 Linch DC, Rode& CH, Jones HM, Brent L. Attempted bone marrow transplantation in a 17-week fetus. Lancet 2:1453, 1986.

[I991 Touraine JL, Raudrant D, Royo C et al. In utero transplanta- tion of stem cells in bare lymphocyte syndrome. Lancet I: 1382, 1989.

[200] Touraine JL, Raudrant D, Vullo C et al. New developments in stem cell transplantation with special reference to the first in utero transplants in humans. Bone Marrow Transplant 7:92-97, 1991.

[201] Touraine JL, Raudrant D, Rebaud A et al. In utero transplanta- tion of stem cells in humans: immunological aspects and clinical follow-up of patients. Bone Marrow Transplant 9: 121-126, 1992.

[202] Raudrant D, Touraine JL, Rebaud A. In utero transplantation of stem cells in humans: technical aspects and clinical experience during pregnancy. Bone Marrow Transplant 9:98-100, 1992.

[203] Bernstein A, Berger D, Hyszar D, Dick J. Gene transfer with retrovirus vectors. In: Setlon JK, Hooaender A, eds. Genetic Engineering: Principles and Methods, Vol. 7. New York: Plenum Press, 1985; 235.

VW Friedmann T. Progress toward human gene therapy. Science 244:1275-1281, 1989.

[205] Apperley JF, Williams DA. Gene therapy. Current status and future directions. Br J Haematol 75:148-155, 1990.

[206] Miller AD. Progress toward human gene therapy. Blood 76271-278, 1990.

[207] Chang JM, Johnson GR. Gene transfer into hemopoietic stem cells using retroviral vectors. Int J Cell Clon 7:264-280, 1989.

[208] Williams DA. Expression of introduced genetic sequences in hematopoietic cells following retroviral-mediated gene transfer. Hum Gene Therap 1:229-239, 1990.

(2091 Karlsson S. Treatment of genetic defects in hematopoietic cell function by gene transfer. Blood 78:2481-2492, 1991.

[210] Lucarelli G, Polchi P, Galimberti M et al. Marrow transplanta- tion for thalassaemia following busulphan and cyclophospha- mide. Lancet 1:1355-1357, 1985.

I2111 Markert ML, Her&field MS, Schiff RI, Buckley RH. Adeno- sine deaminase and purine nucleoside phosphorylase deticien- ties: evaluation of therapeutic interventions in eight patients. J Clin Immunol 7:389-399, 1987.

[212] Foroozanfar N, Hobbs JR, Hugh-Jones K et al. Bone marrow transplantation from an unrelated donor for chronic granulomatous disease. Lancet 1:210-213, 1977.

[2 131 Fischer A, Trung PH, Descamps-Latscha B et al. Bone marrow transplantation for inborn error of phagocytic cells associated with defective adherence, chemotaxis, and oxidative response during opsonized paricle phagocytosis. Lancet 2:473-476,1983.

12141 Hobbs JR, Hugh-Jones K, Shaw PJ, Lindsay I, Hancock M. Beneficial effect of pre-transplant splenectomy on displacement bone marrow transplantation for Gaucher’s syndrome. Lancet 1:1111-1115, 1987.

12151 Vellodi A, Hobbs JR, O’Donnell NM, Coulter BS, Hugh-Jones

K. Treatment of Niemann-Pick disease type B by allogeneic bone marrow transplantation. Br Med J 295:1375-1376, 1987.

[216] Krivit W, Shapiro E, Kennedy Wet al. Treatment of late infan- tile metachromatic leukodystrophy by bone marrow transplan- tation. N Engl J Med 322:28-32, 1990.

I2171 Rosenberg SA. The immunotherapy and gene therapy of cancer. J Clin Oncol 10:180-199, 1992.

[218] Williams DA, Moritz T. Retroviral mediated gene transfer of adenosine deaminase. Exp Hematol21:lOlta, 1993.

[219] Valerio D, Van Beusechem VW, Einerhand MPW, Hooger- brugge PM, Kaptein L. Stem cell gene therapy for adenosine deaminase deficiency. Exp Hematol 21:lOl la, 1993.

[220] Williams DA, Orkin SH, Mulligan RC. Retrovirus-mediated transfer of human adenosine deaminase gene sequences into cells in culture and into mtnine hematopoietic cells in vivo. Proc Nat1 Acad Sci USA 83:2566-2570, 1986.

I2211 Lim B, Williams DA, Orkin SH. Retrovirus-mediated gene transfer of human adenosine deaminase: expression of function- al enzyme in murine hematopoietic stem cells in vivo. Mol Cell Biol 7:3459-3465, 1987.

[222] Lim B, Apperley JF, Orkin SH, Williams DA. Long-term ex- pression of human adenosine deaminase in mice transplanted with retrovirus-infected hematopoietic stem cells. Proc Nat1 Acad Sci USA 868892-8896, 1989.

12231 Wilson JM, Danos 0, Grossman M, Raulet DH, Mulligan RC. Expression of human adenosine deaminase in mice reconstituted with retrovims-transduced hematopoietic stem cells. Proc Nat1 Acad Sci USA 87~439-443, 1990.

I2241 Einerhand MPW, Bakx TA, Kukler A, Valerio D. Factors effec- ting the transduction of pluripotent hematopoietic stem cells: long-term expression of a human adenosine deaminase gene in mice. Blood 81:254-263, 1993.

[225) Kantoff PW, Gillio AP, McLachhn JR et al. Expression of human adenosine deaminase in non-human primates after retrovirus-mediated gene transfer. J Exp Med 166:219-234, 1987.

[226] Bodine DM, Moritz T, Donahue RE et al. Long-term in vivo ex- pression of a murine adenosine deaminase (ADA) gene in rhesus monkey hematopoietic cells of multiple lineages after retroviral mediated gene transfer into CD34+ bone marrow cells. Blood 82:1975-1980, 1993.

[227] Kohn DB, Weinberg KI, Nolta JA et al. Engraftment of gene- modified umbilical cord blood cells in neonates with adenosine deaminase deficiency. Nature Medicine 10: l-7, 1995.

[228] Nimgaonkar M, Bahnson A, Boggs S et al. G-CSF primed peripheral blood stem cells as targets for gene therapy in Gaucher disease. Exp Hematol 21:116Oa, 1993.

[229] Xu L, Stahl SK, Dave HPG et al. Correction of the enzyme de& ciency in hematopoietic cells of Gaucher patients using a clinically acceptable retroviral supematant transduction proto- col. Exp Hematol 22:223-230, 1994.

[230] Karlsson S, Correll PH, Xu LC. Gene transfer and bone marrow transplantation with special reference to Gaucher’s disease. Bone Marrow Transplant 11:124-127, 1993.

[231] Fink JK, Correll PH, Perry LK, Brady RO, Karlsson S. Correc- tion of ghtcocerebrosidase deficiency after retroviral-mediated gene transfer into hematopoietic progenitor cells from patients with Gaucher disease. Proc Nat1 Acad Sci USA 87:2334-2338, 1990.

[232] Morgan RA, Ragheb J, Chuah-VandenDriessche M, Vanden- Driessche T, Dettenbhofer M, Bressler P. Progress towards gene therapy for AIDS. Exp Hematol 21:1012a, 1993.

[233] Yu M, Poeschla E, Wong-Staal F. Progress towards gene thera- py for HIV infection. Gene Ther 1:13-26, 1994.

[234] Moritz T, Keller DC, Williams DA. Human cord blood cells as targets for gene transfer. Potential use in genetic therapies of se-

Page 18: Stem cells from bone marrow, umbilical cord blood and peripheral blood for clinical application: current status and future application

78 L. Lu et al. /Critical Reviews in Oncology/Hematology 22 (19%) 61-78

vere combined immunodeficiency disease. J Exp Med 178529-536, 1993.

12351 Lu L, Kiao M, Clapp DW, Li Z-H, Broxmeyer HE. High etli- ciency retroviral mediated gene transduction into single isolated immature and replatable CD343+ hematopoietic stem/pro- genitor cells from human umbilical cord blood. J Exp Med 178:2089-2096, 1993.

[236] Gruber HE, Finley KD, Hershberg RM et al. Retroviral vector- mediated gene transfer into human hematopoietic progenitor cd. Science 230:1057-1061, 1985.

[237] Hock RA, Miller AD. Retrovirus-mediated transfer and expres- sion of drug resistance genes in human hematopoietic pro- genitor cells. Nature 320:275-277, 1986.

[238] Hogge DE, Humphries RK. Gene transfer to primary normal and malignant human hemopoietic progenitors using recombi- nant retroviruses. Blood 6961 I-617, 1987.

(2391 Nolta JA, Kohn DB. Comparison of the effects of growth fac- tors on retroviral vector-mediated gene transfer and the prolifer- ative status of human hematopoietic progenitor cells. Hum Gene Therap 1:257-268, 1990.

[240] Nolta JA, Crooks GM, Overell RW, Williams DE, Kohn DB. Retroviral vector-mediated gene transfer into primitive human hematopoietic progenitor cells: effects of mast cell growth factor (MGF) combined with other cytokines. Exp Hematol (NY) 20:1065-1071, 1992.

[241] Lu L, Xiao M, Clapp DW, Li Z-H, Broxmeyer HE. Stable inte- gration of retrovirally transduced genes into human umbilical cord blood high proliferative potential colony forming cells (HPP-CFC) as assessed after multiple HPP-CFCcolony replatings in vitro. Blood Cells 20: 525-530, 1994.

[242] Shj Y-J, Shen R-N, Lu L, Broxmeyer HE. Comparative analysis

of transducing genes into cord blood progenitors with retroviral vectors in the presence and absence of growth factors. Blood Cells 20:517-524, 1994.

[243] Moth T, Williams DA. Gene transduction of hematopoietic cells. Focus Growth Factors 45-9, 1993.

Biographies

Li Lu, M.D. is a Full Scientist of Medicine and a Full Member of the Walther Oncology Center, Indiana Uni- versity School of Medicine, Indianapolis, Indiana, USA. She is presently Corresponding Deputy Editor (China) for the Journal, Experimental Hematology. Rang-Nian Shen, M.D. was a Full Scientist of Medicine and Radia- tion Biology and a Full Member of the Walther On- cology Center, Indiana University School of Medicine, Indianapolis, Indiana, USA. He is now retired and resides in China. Hal E. Broxmeyer, Ph.D. is the Mary Margaret Walther Professor of Medicine, Professor of Microbiology and immunology and the Scientific Direc- tor and a Full Member of the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana, USA. He is a former President of the Intema- tional Society for Experimental Hematology and served on numerous national and international committees and editorial boards.