ArticlePDF AvailableLiterature Review

Peripheral Blood Stem Cells for Allogeneic Transplantation: A Review

Authors:
  • Dana-Farber Cancer Institute/Brigham and Women's Hospital

Abstract and Figures

Peripheral blood stem cells (PBSCs) have become increasingly popular for use in hematopoietic stem cell transplantation. PBSCs are readily collected by continuous-flow apheresis from patients and healthy donors after the administration of s.c. recombinant colony-stimulating factors with only minimal morbidity and discomfort.Although the precise identification of PBSCs remains elusive, they can be phenotypically identified as a subset of all circulating CD34+ cells. There are important phenotypic and biologic distinctions between PBSCs and bone marrow (BM)-derived progenitor cells. PBSCs express more lineage-specific antigens but are less metabolically active than their BM-derived counterparts.The use of PBSCs for allogeneic transplantation has been compared to BM in several randomized trials and cohort studies. The use of PBSCs in leukemia, myeloma, non-Hodgkin's lymphoma, and myelodysplasia has resulted in shorter times to neutrophil and platelet engraftment at the expense of increased rates of chronic graft-versus-host disease. The increase in graft-versus-host disease is mainly due to a log-fold increase in donor T cells transferred with the graft. Relapse rates after transplantation may be lower after PBSC transplantation but a convincing survival advantage has not been demonstrated overall. It is possible that a stronger graft-versus-tumor effect may exist with PBSCs when compared with BM although the mechanisms leading to this effect are not clear.
Content may be subject to copyright.
DOI: 10.1634/stemcells.19-2-108
2001;19;108-117 Stem Cells
Corey Cutler and Joseph H. Antin Peripheral Blood Stem Cells for Allogeneic Transplantation: A Review
This information is current as of August 30, 2007
http://www.StemCells.com/cgi/content/full/19/2/108
located on the World Wide Web at:
The online version of this article, along with updated information and services, is
1066-5099. Online ISSN: 1549-4918.
Durham, North Carolina, 27701. © 2001 by AlphaMed Press, all rights reserved. Print ISSN:
Journal is owned, published, and trademarked by AlphaMed Press, 318 Blackwell Street, Suite 260,
STEM CELLS® is a monthly publication, it has been published continuously since 1983. The
genetics and genomics; translational and clinical research; technology development.
embryonic stem cells; tissue-specific stem cells; cancer stem cells; the stem cell niche; stem cell
STEM CELLS®, an international peer-reviewed journal, covers all aspects of stem cell research:
by on August 30, 2007 www.StemCells.comDownloaded from
Peripheral Blood Stem Cells for Allogeneic Transplantation: A Review
COREY CUTLER, JOSEPH H. ANTIN
Department of Adult Oncology, Dana-Farber Cancer Institute and Department of Medicine,
Brigham and Women’s Hospital, Boston, Massachusetts, USA
Key Words. Peripheral blood stem cell · CD34+· Allogeneic · Transplantation · Hematologic malignancies · Apheresis
ABSTRACT
Peripheral blood stem cells (PBSCs) have become
increasingly popular for use in hematopoietic stem cell
transplantation. PBSCs are readily collected by contin-
uous-flow apheresis from patients and healthy donors
after the administration of s.c. recombinant colony-
stimulating factors with only minimal morbidity and
discomfort.
Although the precise identification of PBSCs remains
elusive, they can be phenotypically identified as a subset of
all circulating CD34+cells. There are important pheno-
typic and biologic distinctions between PBSCs and bone
marrow (BM)-derived progenitor cells. PBSCs express
more lineage-specific antigens but are less metabolically
active than their BM-derived counterparts.
The use of PBSCs for allogeneic transplantation has
been compared to BM in several randomized trials and
cohort studies. The use of PBSCs in leukemia, myeloma, non-
Hodgkin’s lymphoma, and myelodysplasia has resulted in
shorter times to neutrophil and platelet engraftment at the
expense of increased rates of chronic graft-versus-host dis-
ease. The increase in graft-versus-host disease is mainly due
to a log-fold increase in donor T cells transferred with the
graft. Relapse rates after transplantation may be lower after
PBSC transplantation but a convincing survival advantage
has not been demonstrated overall. It is possible that a
stronger graft-versus-tumor effect may exist with PBSCs
when compared with BM although the mechanisms leading
to this effect are not clear. Stem Cells 2001;19:108-117
STEM CELLS 2001;19:108-117 www.StemCells.com
Correspondence: Joseph H. Antin, M.D., Dana-Farber Cancer Institute, 44 Binney St., Boston, Massachusetts 02115, USA.
Telephone: 617-632-2525; Fax: 617-632-5175; e-mail: Joseph_Antin@dfci.harvard.edu Received December 21, 2000;
accepted for publication January 8, 2001. ©AlphaMed Press 1066-5099/2001/$5.00/0
INTRODUCTION
Since the first report detailing the use of peripheral
blood-derived stem cells (PBSCs) was published [1], there
has been rapid expansion in the clinical use of these cells as
well as a concomitant increase in an understanding of their
basic biology.
PBSC transplantation (PBSCT) has become increas-
ingly common in the autologous setting, with PBSC largely
replacing bone marrow (BM) as the preferred stem cell
source due largely to quicker engraftment kinetics and ease
of collection. The use of PBSC in allogeneic transplantation
has increased greatly as well, albeit more cautiously.
In this review, we will discuss the identification and
immunophenotypic characteristics of PBSCs, describe
methods for collection of these cells and discuss outcome
issues such as engraftment kinetics and graft-versus-host
disease (GVHD). The clinical experience of PBSCT in var-
ious hematological malignancies with both traditional and
nonmyeloablative approaches will be discussed in detail.
IDENTIFICATION OF PBSCS
Hematopoietic stem cells are normally found in very
limited numbers in the peripheral circulation (less than
0.1% of all nucleated cells). It is logical that progenitor
Stem Cells
Concise Review
by on August 30, 2007 www.StemCells.comDownloaded from
Cutler, Antin
109
cells circulate in the periphery, as this ensures an even dis-
tribution of hematopoiesis within the BM. The movement
of PBSCs in the circulation and homing of PBSCs to the
BM are beyond the scope of this review.
PBSCs represent a subpopulation of all CD34+cells found
in the circulation. Although morphologically difficult to iden-
tify, these cells can be distinguished to some degree by their
immunophenotypic patterns. PBSCs are CD34+/CD38and do
not express a full complement of either myeloid or lymphoid
lineage-specific markers (Lin) but do express the Thy-1
differentiation antigen. These CD34+/CD38/Lin/Thy-1+
cells are the cells responsible for initiating long-term culture
initiating colony (LTC-IC) assays [2].
PBSCs that are mobilized by colony-stimulating factors
(i.e., recombinant human [rHuG-CSF]) are neither phenotyp-
ically nor immunologically identical to BM-derived stem
cells. In comparison with BM-derived stem cells, mobilized
PBSCs have been found to express more lineage-specific dif-
ferentiation antigens (i.e., CD13, CD33), have a lower pro-
portion of cells in S phase (i.e., are less active in cellular
cycling) and are less metabolically active (in rhodamine
retention assays and by demonstrating less CD71 positivity)
[3]. Furthermore, PBSCs demonstrate higher clonogenicity
in LTC assays [3].
Currently, the most reproducible method of stem cell
quantification after collection is by flow cytometric evalua-
tion of CD34+cell numbers. Enumeration of CD34+/CD38,
CD34+/CD33, and CD34+/Thy-1+cell subsets may prove
to be a more useful technique of estimation of stem cell
numbers [4]. The percentage of colony-forming units-gran-
ulocyte-macrophage from a stem cell harvest has also been
used to estimate stem cell numbers. This method is much
less reliable and can vary widely due to differences in cul-
ture technique and media as well as several human factors.
Furthermore, not all transplantation centers are equipped to
perform these labor-intensive cultures, which require 10 to
14 days of incubation. The standardized method for enu-
merating CD34+cell counts has been published in greater
detail elsewhere [2, 5].
MOBILIZATION AND COLLECTION OF PBSCS
Levels of pluripotent hematopoietic stem cells rise up to
50-fold in the recovery phase after myelosuppressive
chemotherapy and can be collected for autologous trans-
plantation. In order to achieve circulating levels high enough
to ensure a harvest capable of reconstituting a mature
hematopoietic system after allogeneic donation, healthy
donors must be primed with hematopoietic growth fac-
tors, using either rHuG-CSF or rHuGM-CSF. The use of
growth factors during the recovery phase after chemother-
apy may or may not increase the total number
of PBSCs that can be collected in the autologous setting.
Combined priming with both nonmyeloablative chemo-
therapy and CSF is not routinely performed in healthy donors.
CSF doses of between 2 and 24 µg/kg administered s.c.
daily have been given to healthy donors [6-8], including
donors over the age of 60 years [9]. The induced leucocy-
tosis, when maintained at levels below 70,000 cells/µl, has
not been shown to be detrimental to the donors health for
short periods of time, however, important morbidity,
including splenic rupture and death, have rarely been
reported [10, 11]. Common minor side effects caused by the
administration of growth factors include bone pain, myal-
gias, headache, and fever, which respond to mild analgesics
in over 80% of cases. Longer follow-up (up to six years)
has confirmed the safety of administration of rHuG-CSF to
healthy donors [11, 12].
PBSCs are obtained by apheresis, generally via periph-
eral venous access; occasionally central venous access may
be required. Up to two to three donor blood volumes are
processed per session by extracorporeal continuous-flow
apheresis machines. Stem cells and granulocytes are sepa-
rated from the red blood cell and plasma fractions of blood
by centrifugation and the latter two components are returned
to the donors during the apheresis procedure itself. The
process generally requires between three and five hours per
session. The target range of peripheral stem cells of 2 ×106
cells/kg (recipient weight) can generally be achieved in one to
two sessions.
The efficacy of stem cell collection comparing BM and
PBSCs has been examined. In a randomized trial, a single
PBSC apheresis procedure yielded 3.7 times more CD34+
cells than a standard BM harvest. Furthermore, BM har-
vests were 6.8 times more likely to be insufficient for trans-
plantation (defined as less than 2 ×106cells/kg of recipient
weight, p< 0.001) [13].
Potential risks associated with stem cell apheresis proce-
dure include complications related to central line placement
when necessary (such as pneumothorax) and cytopenias due
to the apheresis procedure itself. Leukopenia or lymphope-
nia frequently occur and can last a variable period of time.
Thrombocytopenia (<100 ×109cells/l) is not uncommon,
however, bleeding complications are extraordinarily rare.
Similarly, anemia that requires transfusion is very uncom-
mon. Anderlini et al. provide a complete review of donor
safety issues [14].
PBSC donation is generally preferred by donors over
traditional BM donation, which entails either general or
spinal anesthesia, a brief hospital stay, and more post-proce-
dure discomfort. Several phase II studies measuring outcomes
(including quality of life and anxiety scores) comparing the
two procurement procedures confirmed this fact [15, 16].
by on August 30, 2007 www.StemCells.comDownloaded from
110
PBSCs for Allogeneic Transplantation
ENGRAFTMENT AND KINETICS
The prolonged and profound cytopenias encountered
after transplantation account for much of the morbidity and
mortality associated with the procedure. Typical delays to
neutrophil recovery after autologous BM transplantation
(BMT) range from 14 to 21 days. After allogeneic BMT, the
time to engraftment is longer and more variable, ranging from
14 to 28 days and in part may be related to post-transplant
immunosuppressive agents, such as methotrexate.
A convincing reduction in time to engraftment after both
autologous and allogeneic PBSCT when compared to tradi-
tional BMT has been demonstrated. In the allogeneic setting,
neutrophil engraftment (to 0.5 ×109cells/l on two consecutive
days) occurred between one and six days earlier with PBSCT
when compared with BMT in randomized trials (mean time to
engraftment: 14 versus 15 days and 15 versus 21 days) [17,
18]. Unsupported platelet counts of 20 ×109/l occurred
between four and seven days earlier (mean time to platelet
engraftment: 15 versus 19 days and 11 versus 18 days) [17,
19]. The results of a large database review are consistent with
the results of the randomized trials (mean time to neutrophil
engraftment 14 versus 19 days, mean time to platelet engraft-
ment 18 versus 25 days, p< 0.001 for both comparisons) [20].
Typical doses of CD34+stem cells used for PBSCs are
2×106cells/kg of recipient body weight or greater. Doses
lower than this threshold are associated with prolonged
cytopenias and increased early mortality [21].
A relationship between the dose of CD34+stem cells
delivered with the transplant and the tempo of hematologic
recovery has been demonstrated for both BMT [21] as well
as PBSCT [22]. The use of higher doses of CD34+cells may
lead to quicker engraftment, particularly when doses are
greatly increased [23, 24]. Platelet recovery appears to be
more sensitive to CD34+doses than neutrophil recovery [24].
Efforts to enrich PBSCT by ex-vivo CD34+cell selection
(positive selection) have resulted in increased rates of GVHD,
possibly by altering the cytokine expression patterns of trans-
planted cells or changing lymphocyte subsets delivered with
the graft [25]. These efforts do not appear to alter engraftment
kinetics significantly [25, 26]. Negative selection (T cell
depletion) clearly leads to lower rates of GVHD, at the
expense of a slightly higher incidence of graft rejection.
Other strategies used to shorten the time to engraftment
include the use of combined PBSCT and BMT [27] and the
use of rHuG-CSF-mobilized BM for transplantation [28-30].
The latter approach may have the advantage of GVHD rates
similar to traditional BMT [29] and neutrophil engraftment
kinetics similar to those of PBSCT [30].
The earlier engraftment seen after PBSCT has lead to
earlier discharge from hospital [17, 19, 31] and total lower
immediate costs associated with the transplant procedure
[18, 32]. The reduction in costs associated with the proce-
dure is primarily due to fewer dollars spent on hospital room
charges, blood products and other supportive measures. The
costs of stem cell mobilization and collection procedures,
however, are greater for PBSCT than for traditional BMT,
primarily due to the use of recombinant human hematopoi-
etic growth factors [32]. Long-term cost issues are more dif-
ficult to predict and will be influenced by GVHD outcomes
after PBSCT (see below).
GVHD
Acute GVHD results from the complex interaction of
donor T cells and recipient organs and involves recogni-
tion of minor histocompatibility antigens in an inflamma-
tory milieu. Tissue damage from conditioning regimens,
infections, and the underlying illness may provide an envi-
ronment that fosters T cell recognition of susceptible tis-
sues. Clinical injury is thought to derive from direct T cell
injury through perforin/granzyme, Fas/Fas ligand interac-
tions, and the effects of inflammatory cytokines [33]. It
may be due in part to conditioning-induced tissue damage,
transmigration of endotoxin across damaged gut mucosa,
and dysregulation of inflammatory cytokines.
Acute GVHD occurs within the first weeks following
transplantation. Chronic GVHD occurs later, and is arbitrar-
ily defined as the presence or persistence of GVHD beyond
100 days since transplantation. The occurrence and severity
of GVHD is related to the degree of HLA-mismatching
between donor and recipient, the type of conditioning regi-
men used, the use of degree of immunosuppression of the
recipient (GVHD prophylaxis), the viral exposure of the
donor and recipient, the underlying malignant disease, and
to the passage of mature, immunocompetent T cells with the
stem cell transplant [34]. PBSCT contain roughly a log-fold
increase in the concentration of CD3+T cells when com-
pared to BMT. This large increase in T cell dose is largely
responsible for the increased risk of GVHD seen after
PBSCT. PBSCs were avoided for years because of concerns
over the higher risk of GVHD, however, these concerns
appear to be less severe than originally anticipated.
The incidence and severity of GVHD after PBSCT may
be influenced by rHuG-CSF administered to donors prior to
stem cell collection. Polarization of T cells to the Th2 class
has been demonstrated to occur in response to rHuG-CSF in
mouse models [35, 36]. This polarization is related to the
subtype of antigen-presenting cell (dendritic cell) initiating
the immune response. rHu-G-CSF has been shown to
increase the circulating numbers of type 2 dendritic cells,
which in turn influence the predominance of a Th2 cellular
response and elucidated cytokines [37, 38]. Th2 cells are
considered GVHD neutral in comparison to Th1 cells.
by on August 30, 2007 www.StemCells.comDownloaded from
Cutler, Antin
111
There have been five randomized controlled trials [17-
19, 39, 40] and many cohort studies [20, 31, 41-50] that
have examined the relative risks (RR) of acute and chronic
GVHD after PBSCT when compared to traditional BMT
(Table 1). No single trial has concluded that the incidence of
acute GVHD incidence is higher after PBSCT when com-
pared to traditional BMT. Only one of the trials, however,
was statistically powered to detect small differences in the
incidence of acute GVHD [40]. RR for acute GVHD after
PBSCT varied from 0.67 to 1.5 compared to BMT [43, 44].
In a meta-analysis of 15 trials, a small but statistically sig-
nificant increase in the RR of acute GVHD after PBSCT
was demonstrated (RR 1.13, 95% confidence interval [CI]
1.01-1.26) [51].
It has been accepted clinically that the incidence of
chronic GVHD after PBSCT may be higher than after tra-
ditional BMT. The literature regarding this subject has been
controversial, with some studies finding evidence support-
ing [18, 20, 49, 50] and others not supporting this hypothe-
sis [19, 39, 40]. In a meta-analysis, a 1.5-fold increase in
RR was noted when the results of 13 trials were combined
(RR 1.48, 95% CI 1.20-1.81) [51]. In addition to being
more prevalent after PBSCT, chronic GVHD was more
likely to be extensive when compared with BMT (RR 1.58,
95% CI 1.24-2.00) [51]. This increase in chronic GVHD
was related to the increased dose of T cells delivered with
the graft in a regression model [51].
The occurrence of acute GVHD has been linked to higher
treatment-related morbidity and early mortality, however,
chronic GVHD has been shown to be protective against
relapse [51, 52] and may be associated with better long-term
survival, particularly when used in high-risk patients [20, 40].
It remains to be determined whether the reduction in hospital
stay during the acute phase is cost-effective compared with
the increased costs and morbidity of chronic GVHD.
USE OF PBSCS FOR SPECIFIC HEMATOLOGICAL
MALIGNANCIES
Autologous stem cell grafting has been used with vary-
ing degrees of success in chronic myelogenous leukemia
(CML) [53, 54], acute leukemia [55], myelodysplasia [56],
and multiple myeloma [57]. The potential advantages of allo-
geneic transplantation include the security of a contaminant-
free graft and the possibility of a unique graft-versus-tumor
effect. The use of PBSC when compared to BM may lead to
a reduction in relapse rates after transplantation, due to an
enhanced graft-versus-tumor effect. An overall improvement
in survival has not been demonstrated, however, there may
be a trend towards better outcomes after PBSCT in patients
with high-risk disease.
Acute and Chronic Leukemia
BMT for acute myelogenous leukemia (AML) is recom-
mended for patients with refractory/resistant leukemia,
patients in second clinical remission, and patients with high-
risk cytogenetic features in first remission. The Medical
Research Council AML10 trial revealed significant differ-
ences in long-term disease-free survival when autologous
Table 1. RR of acute and chronic GVHD after PBSCT versus BMT
Study Study design nRR of acute RR of chronic
GVHD (95% CI) GVHD (95% CI)
Powles [19] Single institution blind randomized 39 1.06 (0.55-2.01) 1.52 (0.60-3.83)
Blaise [18] Multicenter randomized 101 1.06 (0.67-1.66) 1.82 (1.10-3.00)
Bensinger [40] Multicenter randomized 172 1.19 (0.92-1.54) 1.19 (0.85-1.67)
Vigorito [39] Single institution randomized 37 1.42 (0.38-5.33) 1.34 (0.75-2.39)
Schmitz [17] Multicenter randomized 66 1.13 (0.70-1.80) 1.50 (0.38-6.00)
Champlin [20] Retrospective database cohort 824 1.19 (0.91-1.56) 1.30 (1.00-1.70)
Russell [46] Retrospective cohort 87 1.50 (0.75-3.00) 1.76 (1.26-2.46)
Üstün[50] Cohort with historical controls 80 1.03 (0.51-2.06) 3.65 (1.75-7.58)
Scott [47] Cohort with historical controls 24 1.00 (0.39-2.58) 3.33 (0.47-23.47)
Solano [48] Retrospective cohort 74 NA 1.89 (0.97-3.68)
Bacigalupo [42] Retrospective cohort 97 1.25 (0.85-1.84) 0.99 (0.90-1.09)
Lemoli [44] Cohort with historical controls 30 1.50 (0.29-7.73) 1.40 (0.57-3.43)
Pavletic [45] Cohort with historical controls 43 1.20 (0.67-2.15) NA
Bensinger/Storek [43, 49] Cohort with historical controls 74 0.67 (0.40-1.13) 1.67 (1.09-2.54)
Azevedo [41] Cohort with historical controls 38 1.11 (0.59-2.10) NA
Study, year of publication = the first author and the year of publication; n= study sample size; 95% CI = 95% confidence intervals for RR assessments;
RR = relative risk.
by on August 30, 2007 www.StemCells.comDownloaded from
112
PBSCs for Allogeneic Transplantation
transplantation was compared with consolidative chemother-
apy in individuals without an HLA-matched sibling [55].
At least one trial has demonstrated higher long-term sur-
vival after allogeneic transplantation compared with autol-
ogous transplantation [58], but others have demonstrated
equivalence [57, 59]. Allogeneic transplantation has
become the standard of care for patients with high-risk
cytogenetic features in first remission, patients with resis-
tant or refractory disease and patients in second remission,
provided the patient is a candidate for the procedure and a
suitable donor is available. Acute lymphoblastic leukemia
is treated largely the same way in the adult population.
In stable phase CML, allogeneic transplantation is cur-
rently the curative therapy of choice when feasible.
Autologous transplantation with Philadelphia chromosome-
negative-purged marrow or stem cells has recently re-
emerged as an investigational treatment option [60], but
there is no established benefit of autologous transplantation
over interferon-based therapy.
PBSCT has been used most extensively in acute and
chronic leukemias. In a prospective, randomized trial com-
paring allogeneic BMT and PBSCT in 111 patients, Blaise
et al. did not demonstrate significantly different overall and
leukemia-free survival between PBSCT and BMT groups
[18]. The trial did demonstrate the expected significantly
decreased time to neutrophil and platelet engraftment after
PBSCT. The rate of chronic GVHD was noted to be signif-
icantly higher after PBSCT when compared with BMT
(30% versus 55%, p< 0.03).
In a study reported by Schmitz et al., the authors reported
the outcomes of 66 patients with acute or chronic leukemia
who received BMT or PBSCT from HLA-matched siblings
[17]. The majority of patients entered in this trial had low-risk
disease, referring to AML in first remission or CML in
chronic, stable phase. The mean time to platelet recovery (>20
×109/l) was reduced by four days after PBSCT when com-
pared to BMT. No differences in rates of GVHD or overall
survival were reported in this trial.
Multiple Myeloma
Autologous BMT for multiple myeloma has been
shown to be superior to traditional chemotherapy, offering
both a disease-free and overall survival advantage [61].
However, autologous transplants are frequently contami-
nated by residual tumor cells when assessed by sensitive
molecular techniques [53, 62]. The risk of contamination as
well as the demonstration of a pronounced graft-versus-
myeloma effect [63, 64] has prompted the study of allo-
geneic stem cell sources as an alternative to autologous
cells. This approach is not without risk; the Cancer and
Leukemia Group B recently has had to stop accrual to a trial
of non-T cell-depleted allogeneic transplantation in myeloma
due to concerns over toxicity.
Majolino et al. described 10 patients (including four who
had already received PBSC autografts) undergoing allogeneic
PBSCT for multiple myeloma [65]. The median time to
engraftment was 13 days. Eight of 10 patients had a complete
remission (CR) while the remaining two patients had a partial
remission (PR). None of the patients achieving CR had evi-
dence of recurrence of myeloma 7 to 28 months after trans-
plantation. Two patients died of treatment-related causes.
Corradini et al. described the molecular remission status
of 51 patients after autografting or allogeneic transplantation
with either BM or PBSCs [66]. All 17 patients who received
allografts entered a CR state after transplantation. In patients
with molecular markers available, there was a significantly
increased proportion of patients who achieved a molecular
remission after allogeneic transplantation compared to
autologous transplantation. Furthermore, only one of five
allogeneic BMT patients became polymerase chain reac-
tion-negative for clonal immunoglobulin gene rearrange-
ments compared with six of nine patients who received
allogeneic PBSCT. Similarly, Cavo et al. described five
patients who had both complete clinical and molecular
remissions after PBSCT from HLA-matched siblings [67].
Myelodysplasia
Allogeneic transplantation is the only therapy for
myelodysplastic syndromes (MDS) with curative potential
available today. Patients with fewer cytogenetic abnormali-
ties [68], less latency since the time of diagnosis [69], and
patients with refractory anemia or refractory anemia with
ringed sideroblasts [70, 71] have better outcomes after trans-
plantation. The International Prognostic Scoring System
score is also useful for predicting outcome after transplanta-
tion [72]. The optimal timing of stem cell transplantation for
this disease is not yet known.
The experience in transplantation with PBSC for MDS
is more limited than that for the acute and chronic
leukemias. Patients with MDS have comprised only a small
proportion of patients in trials comparing PBSCT and BMT
and therefore no conclusions can be drawn on the relative
advantages and disadvantages of PBSCT.
Non-Hodgkins Lymphoma (NHL)
Autologous transplantation for relapsed intermediate and
high-grade NHL has clear survival advantages over
chemotherapy [73]. Allogeneic transplantation has been used
for patients with relapse after BMT or refractory disease. In a
preliminary report by Körbling et al., four patients with
refractory NHL underwent allogeneic PBSCT from HLA-
matched siblings [74]. Three patients had a CR and the fourth,
by on August 30, 2007 www.StemCells.comDownloaded from
Cutler, Antin
113
a PR. One patient died of infectious complications 82 days
after transplantation and the other three patients were reported
to be alive greater than 31 to 100 days post-transplant.
Khouri et al. studied the effects of allogeneic transplan-
tation in patients with mantle cell lymphoma, an intermedi-
ate-grade lymphoma with a uniformly poor prognosis [75].
Sixteen patients were studied, 11 of whom received allo-
geneic PBSCs as a stem cell source. Nine patients remained
alive at the time of publication, with eight patients in CR.
Molecular minimal residual disease was assessed in seven
patients. Five of the seven patients had no evidence of mole-
cular residual disease between 3 and 30 months after trans-
plantation. Five of seven patients had evidence of chronic
GVHD. Other case reports have documented the efficacy of
allogeneic PBSCT for NHL [76, 77].
A graft-versus-lymphoma effect has been noted in both
animal models [78] and human studies [75-77]. In a murine
model of NHL, Ito and Shizuru demonstrated a unique graft-
versus-lymphoma activity which was separable from GVHD
effects and mediated by CD8+T cells and perforin-dependent
cytolysis [78].
BM Aplasia and Donor Lymphocyte Infusion (DLI)
The use of unstimulated PB buffy coat cells for aplastic
anemia was first reported by Storb et al. in 1982 [79]. At this
time, the addition of buffy coat cells to BMT increased the
risk of GVHD, likely as a result of the immense T cell load
in the buffy coat compared with the relative sparse concen-
tration of progenitor cells. Graft rejection was diminished
and survival for patients treated with both marrow and buffy
coat cells was improved compared with marrow alone; how-
ever, it was not clear whether this improvement could be
attributed to the stem cells infused or prevention of graft
rejection by the infusion of large numbers of allogeneic
T cells [80].
More recently, infusions of buffy coat cells, referred to
as DLI have been used as post-transplantation immunother-
apy for malignant disease. Given at variable times after
transplantation of allogeneic BM or PBSC, DLI can induce
potent graft-versus-tumor effects and is useful as treatment
for relapsed malignancies or pre-emptive therapy for preven-
tion of relapse. The procedure is associated with a 50% risk
in acute GVHD and a 20% risk of marrow aplasia [81]. The
effects of DLI are most prominent in CML, where single
infusions of donor lymphocytes have induced long-lasting
remissions in relapsed patients [82].
Despite the infusion of hematopoietic progenitor cells
with the lymphocyte population, even rHuGM-CSF-mobilized
donor lymphocytes are not completely protective against mar-
row aplasia when used in relapsed CML [81]. This implies that
aplasia seen after DLI may not be solely due to destruction of
native hematopoietic elements, but also involves suppression
of donor engraftment and hematopoiesis.
Nonmyeloablative Transplantation and PBSCs
The DLI experience led to the notion that a transplanta-
tion could rely primarily on the immune system of the
donor to eradicate the leukemia and that sublethal condi-
tioning might allow stable engraftment. Clearly, depending
on whether the targeted disease was a stem cell disorder
(e.g., CML) or a disease in which stem cells are not
involved (e.g., NHL), it may or may not be necessary to
establish complete chimerism of hematopoiesis. The first
trial of nonmyeloablative or reduced-intensity conditioning
followed by allogeneic PBSCT was reported by Khouri et
al. in 1998 [83]. This technique employs the graft-versus-
tumor effect as the primary therapeutic modality and omits
high-dose conditioning regimens. Instead, low-dose
immunosuppressive regimens containing drugs such as the
nucleoside analogue fludarabine, generally in combination
with cyclophosphamide or busulfan, are used to permit
donor progenitor cell engraftment even without host myelo-
ablation. The graft-versus-host/graft-versus-leukemia
response might result in eradication of host hematopoiesis,
therefore it is necessary to include a source of stem cells to
sustain hematopoiesis.
Potential advantages of a nonmyeloablative approach
to allogeneic transplantation include the inclusion of an
older patient population and patients with other comorbid
conditions that would be otherwise excluded from allo-
geneic transplantation. As well, the procedure itself is not
limited by chemotherapy-induced toxicity and can often be
performed in an outpatient setting. Rates of GVHD after
nonmyeloablative transplantation are similar to those seen
after myeloablative transplantation.
In the first published phase II study [83], 15 patients
with lymphoid malignancies were transplanted following
nonmyeloablative conditioning. Eleven of 15 patients
demonstrated evidence of donor-derived hematopoiesis at
one month post-transplant. Mixed chimerism (defined as the
coexistence of recipient and donor-derived hematopoiesis)
was noted in most subjects. DLIs in the post-transplant set-
ting were able to convert some patients from mixed chimeric
hematopoiesis to full donor-derived hematopoiesis.
A nonmyeloablative approach to PBSCT for NHL was
explored by Nagler et al. [84]. Nineteen patients with heav-
ily-pretreated NHL were given a nonmyeloablative condi-
tioning regimen consisting of fludarabine, busulfan, and
antithymocyte globulin. Eight of the 19 patients were alive
between 15 and 37 months post-transplant. There were four
patients who relapsed after treatment. An attempt at post-
transplantation immunomodulation with immunosuppressive
by on August 30, 2007 www.StemCells.comDownloaded from
114
PBSCs for Allogeneic Transplantation
therapy withdrawal and/or DLI was not successful at
inducing remissions in these patients.
CONCLUSIONS AND FUTURE DIRECTIONS
PBSCs are now commonly used as the stem cell source
for allogeneic transplantation. These cells are readily col-
lected from the peripheral circulation when mobilized with
hematopoietic growth factors. Efforts to increase the yield
of donor harvests are now focusing on the optimal timing of
stem cell procurement and on the use of novel hematopoi-
etic growth factors (such as flt-3 ligand) to increase the effi-
cacy of mobilization of the stem cell compartment into the
peripheral circulation.
Although the time to engraftment of PBSCs appears to
be shorter than BM-derived stem cells, newer BM mobiliza-
tion techniques may obviate this difference in the future. The
specific biological factors leading to a more rapid engraft-
ment are yet to be identified fully and represent a gap in the
basic understanding of stem cell biology.
Although the incidence of GVHD was initially thought to
be similar for both BMT and PBSCT, recent evidence from
ongoing clinical trials and a meta-analysis has demonstrated
that both acute and chronic GVHD occur with greater fre-
quency after PBSCT than BMT. Efforts to control GVHD
with novel immunophyllin inhibitors and other immunomodu-
latory agents may reduce rates of GVHD after both procedures
to minimize treatment-related mortality and long-term mor-
bidity. The ability to harness the useful effects of the graft-ver-
sus-tumor activity and separate them from GVHD will further
increase the beneficial effects of high-dose therapy and stem
cell transplantation. The addition of post-transplantation cellu-
lar immunotherapy with either donor lymphocyte or dendritic
cell preparations may enhance graft-versus-tumor activity.
The role of PBSCT for individual malignancies remains
to be determined. At the present, it is reasonable to reserve
the use of PBSC to clinically high-risk scenarios, such as sec-
ond remission AML or CML in blast crisis, where PBSCT
has shown a survival advantage over BMT in a database
review. For low-risk malignant conditions such as stable
phase CML, it is reasonable to continue using BM-derived
stem cells for transplantation until clinical trials demonstrate
the superiority of one stem cell source over the other.
REFERENCES
1 Goldman JM, Thing KH, Park DS et al. Collection, cryo-
preservation and subsequent viability of haemopoietic stem
cells intended for treatment of chronic granulocytic leukaemia
in blast-cell transformation. Br J Haematol 1978;40:185-188.
2 Siena S, Schiavo R, Pedrazzoli P et al. Therapeutic relevance
of CD34 cell dose in blood cell transplantation for cancer
therapy. J Clin Oncol 2000;18:1360-1377.
3 Gyger M, Stuart RK, Perreault C. Immunobiology of allogeneic
peripheral blood mononuclear cells mobilized with granulo-
cyte-colony stimulating factor. Bone Marrow Transplant
2000;26:1-16.
4 To LB, Haylock DN, Simmons PJ et al. The biology and clinical
uses of blood stem cells. Blood 1997;89:2233-2258.
5 Sutherland DR, Anderson L, Keeney M et al. The ISHAGE
guidelines for CD34+cell determination by flow cytometry.
J Hematother 1996;5:213-226.
6 Bensinger WI, Price TH, Dale DC et al. The effects of daily
recombinant human granulocyte colony-stimulating factor
administration on normal granulocyte donors undergoing
leukapheresis. Blood 1993;81:1883-1889.
7 Murata M, Harada M, Kato S et al. Peripheral blood stem cell
mobilization and apheresis: analysis of adverse events in 94
normal donors. Bone Marrow Transplant 1999;24:1065-1071.
8 McCullough J, Clay M, Herr G et al. Effects of granulocyte-
colony-stimulating factor on potential normal granulocyte
donors. Transfusion 1999;39:1136-1140.
9 Anderlini P, Przepiorka D, Lauppe J et al. Collection of periph-
eral blood stem cells from normal donors 60 years of age or
older. Br J Haematol 1997;97:485-487.
10 Becker PS, Wagle M, Matous S et al. Spontaneous splenic rup-
ture following administration of granulocyte colony-stimulating
factor (G-CSF): occurrence in an allogeneic donor of peripheral
blood stem cells. Biol Blood Marrow Transplant 1997;3:45-49.
11 de la Rubia J, Martinez C, Solano C et al. Administration of
recombinant human granulocyte colony-stimulating factor to
normal donors: results of the Spanish National Donor Registry.
Bone Marrow Transplant 1999;24:723-728.
12 Cavallaro AM, Lilleby K, Majolino I et al. Three to six year
follow-up of normal donors who received recombinant
human granulocyte colony-stimulating factor. Bone Marrow
Transplant 2000;25:85-89.
13 Singhal S, Powles R, Kulkarni S et al. Comparison of marrow
and blood cell yields from the same donors in a double-blind,
randomized study of allogeneic marrow vs. blood stem cell
transplantation. Bone Marrow Transplant 2000;25:501-505.
14 Anderlini P, Przepiorka D, Körbling M et al. Blood stem cell
procurement: donor safety issues. Bone Marrow Transplant
1998;21(suppl 3):S35-S39.
15 Auquier P, Macquart-Moulin G, Moatti JP et al. Comparison of
anxiety, pain and discomfort in two procedures of hematopoi-
etic stem cell collection: leukacytapheresis and bone marrow
harvest. Bone Marrow Transplant 1995;16:541-547.
16 Ordemann R, Hölig K, Wagner K et al. Acceptance and fea-
sibility of peripheral stem cell mobilisation compared to bone
marrow collection from healthy unrelated donors. Bone
Marrow Transplant 1998;21(suppl 3):S25-S28.
17 Schmitz N, Bacigalupo A, Hasenclever D et al. Allogeneic
bone marrow transplantation vs. filgrastim-mobilised periph-
eral blood progenitor cell transplantation in patients with early
by on August 30, 2007 www.StemCells.comDownloaded from
Cutler, Antin
115
leukaemia: first results of a randomised multicentre trial of the
European Group for Blood and Marrow Transplantation. Bone
Marrow Transplant 1998;21:995-1003.
18 Blaise D, Kuentz M, Fortanier C et al. Randomized trial of
bone marrow versus Lenograstim-primed blood cell allo-
geneic transplantation in patients with early-stage leukemia: a
report from the Société Française de Greffe de Moelle. J Clin
Oncol 2000;18:537-546.
19 Powles R, Mehta J, Kulkarni S et al. Allogeneic blood and bone-
marrow stem-cell transplantation in haematological malignant
diseases: a randomised trial. Lancet 2000;335:1231-1237.
20 Champlin RE, Schmitz N, Horowitz MM et al. Blood stem
cells compared with bone marrow as a source of hematopoietic
cells for allogeneic transplantation. Blood 2000;95:3702-3709.
21 Mavroudis D, Read E, Cottler-Fox M et al. CD34+cell dose
predicts survival, posttransplant morbidity, and rate of hema-
tologic recovery after allogeneic marrow transplants for
hematologic malignancies. Blood 1996;88:3223-3229.
22 Ilhan O, Arslan Ö, Arat M et al. The impact of the CD34+cell
dose on engraftment in allogeneic peripheral blood stem cell
transplantation. Transfus Sci 1999;20:69-71.
23 Miflin G, Russell NH, Hutchinson RM et al. Allogeneic periph-
eral blood stem cell transplantation for haematological malig-
nanciesan analysis of kinetics of engraftment and GVHD
risk. Bone Marrow Transplant 1997;19:9-13.
24 Shpall EJ, Champlin R, Glaspy JA. Effect of CD34+periph-
eral blood progenitor cell dose on hematopoietic recovery.
Biol Blood Marrow Transplant 1998;4:84-92.
25 Vij R, Brown R, Shenoy S et al. Allogeneic peripheral blood
stem cell transplantation following CD34+enrichment by
density gradient separation. Bone Marrow Transplant
2000;25:1223-1228.
26 Urbano-Ispizua A, Rozman C, Martínez C et al. Rapid engraft-
ment without significant graft-versus-host disease after allo-
geneic transplantation of CD34+selected cells from peripheral
blood. Blood 1997;89:3967-3973.
27 Szer J, Curtis DJ, Bardy PG et al. The addition of allogeneic
peripheral blood-derived progenitor cells to bone marrow for
transplantation: results of a randomised clinical trial. Aust N
Z J Med 1999;29:487-493.
28 Couban S, Messner HA, Andreou P et al. Bone marrow mobi-
lized with granulocyte colony-stimulating factor in related
allogeneic transplant recipients: a study of 29 patients. Biol
Blood Marrow Transplant 2000;6:422-427.
29 Isola L, Scigliano E, Fruchtman S. Long-term follow-up after
allogeneic granulocyte colony-stimulating factor-primed
bone marrow transplantation. Biol Blood Marrow Transplant
2000;6:428-433.
30 Serody JS, Sparks SD, Lin Y et al. Comparison of granulocyte
colony-stimulating factor (G-CSF)-mobilized peripheral blood
progenitor cells and G-CSF-stimulated bone marrow as a
source of stem cells in HLA-matched sibling transplantation.
Biol Blood Marrow Transplant 2000;6:434-440.
31 Przepiorka D, Anderlini P, Ippoliti C et al. Allogeneic blood
stem cell transplantation in advanced hematologic cancers.
Bone Marrow Transplant 1997;19:455-460.
32 Bennett CL, Waters TM, Stinson TJ et al. Valuing clinical strate-
gies early in development: a cost analysis of allogeneic periph-
eral blood stem cell transplantation. Bone Marrow Transplant
1999;24:555-560.
33 Antin JH, Ferrara JL. Cytokine dysregulation and acute graft-
versus-host disease. Blood 1992;80:2964-2968.
34 Przepiorka D, Smith TL, Folloder J et al. Risk factors for
acute graft-versus-host disease after allogeneic blood stem
cell transplantation. Blood 1999;94:1465-1470.
35 Pan L, Delmonte J Jr, Jalonen CK et al. Pretreatment of donor
mice with granulocyte colony-stimulating factor polarizes
donor T lymphocytes toward type-2 cytokine production and
reduces severity of experimental graft-versus-host disease.
Blood 1995;86:4422-4429.
36 Zeng D, Dejbakhash-Jones S, Strober S. Granulocyte colony-
stimulating factor reduces the capacity of blood mononuclear
cells to induce graft-versus-host disease: impact on blood
progenitor cell transplantation. Blood 1997;90:453-463.
37 Arpinati M, Green CL, Heimfeld S et al. Granulocyte-colony
stimulating factor mobilizes T helper 2-inducing dendritic
cells. Blood 2000;95:2484-2490.
38 Reddy V, Hill GR, Pan L et al. G-CSF modulates cytokine
profile of dendritic cells and decreases acute graft-versus-host
disease through effects on the donor rather than the recipient.
Transplant 2000;69:691-693.
39 Vigorito AC, Azevedo WM, Marques JFC et al. A randomized,
prospective comparison of allogeneic bone marrow and periph-
eral blood progenitor cell transplantation in the treatment of
haematological malignancies. Bone Marrow Transplant
1998;22:1145-1151.
40 Bensinger W, Martin P, Clift R et al. A prospective, randomised
trial of peripheral blood stem cells (PBSC) or marrow (BM) for
patients undergoing allogeneic transplantation for hematologic
malignancies. Blood 1999;94(suppl 1):10a.
41 Azevedo WM, Aranha FJP, Gouvea AC et al. Allogeneic
transplantation with blood stem cells mobilized by rhG-CSF
for hematological malignancies. Bone Marrow Transplant
1995;16:647-653.
42 Bacigalupo A, Zikos P, Van Lint M-T et al. Allogeneic bone
marrow or peripheral blood cell transplants in adults with
hematologic malignancies: a single-center experience. Exp
Hematol 1998;26:409-414.
43 Bensinger WI, Clift R, Martin P et al. Allogeneic peripheral
blood stem cell transplantation in patients with advanced
hematologic malignancies: a retrospective comparison with
marrow transplantation. Blood 1996;88:2794-2800.
44 Lemoli RM, Bandini G, Leopardi G et al. Allogeneic peripheral
blood stem cell transplantation in patients with early-phase
hematologic malignancy: a retrospective comparison of short-
term outcome with bone marrow transplantation. Haematologica
1998;83:48-55.
by on August 30, 2007 www.StemCells.comDownloaded from
116
PBSCs for Allogeneic Transplantation
45 Pavletic ZS, Bishop MR, Tarantolo SR et al. Hematopoietic
recovery after allogeneic blood stem-cell transplantation
compared with bone marrow transplantation in patients with
hematologic malignancies. J Clin Oncol 1997;15:1608-1616.
46 Russell JA, Larratt L, Brown C et al. Allogeneic blood stem
cells and bone marrow transplantation for acute myelogenous
leukemia and myelodysplasia: influence of stem cell source
on outcome. Bone Marrow Transplant 1999;24:1177-1183.
47 Scott MA, Gandhi MK, Jestice HK et al. A trend towards an
increased incidence of chronic graft-versus-host disease fol-
lowing allogeneic peripheral blood progenitor cell transplan-
tation: a case controlled study. Bone Marrow Transplant
1998;22:273-276.
48 Solano C, Martinez C, Brunet S et al. Chronic graft-versus-host
disease after allogeneic peripheral blood progenitor cell or bone
marrow transplantation from matched related donors. A case-
control study. Bone Marrow Transplant 1998;22:1129-1135.
49 Storek J, Gooley T, Siadak M et al. Allogeneic peripheral blood
stem cell transplantation may be associated with a high risk of
chronic graft-versus-host disease. Blood 1997;90:4705-4709.
50 Üstün C, Arslan Ö, Beksaç M et al. A retrospective compari-
son of allogeneic peripheral blood stem cell and bone marrow
transplantation results from a single center: a focus on the
incidence of graft-vs.-host disease and relapse. Biol Blood
Marrow Transplant 1999;5:28-35.
51 Cutler C, Giri S, Jeyapalan S et al. Incidence of acute and chronic
graft-versus-host disease after allogeneic peripheral blood stem
cell and bone marrow transplantation: a meta-analysis. Blood
2000;96:205a.
52 Elmaagacli A, Beelen DW, Opalka B et al. The risk of resid-
ual molecular and cytogenetic disease in patients with
Philadelphia-chromosome positive first chronic phase
chronic myelogenous leukemia is reduced after transplanta-
tion of allogeneic peripheral blood stem cells compared with
bone marrow. Blood 1999;94:384-389.
53 McGlave PB, De Fabritiis P, Deisseroth A et al. Autologous
transplants for chronic myelogenous leukaemia: results from
eight transplant groups. Lancet 1994;343:1486-1488.
54 Reiffers J, Goldman J, Meloni G et al. on behalf of the Chronic
Leukemia Working Party of the EBMT. Autologous stem cell
transplantation in chronic myelogenous leukemia: a retrospec-
tive analysis of the European Group for Bone Marrow
Transplantation. Bone Marrow Transplant 1994;14:407-410.
55 Burnett AK, Goldstone AH, Stevens RM et al. Randomised
comparison of addition of autologous bone-marrow transplan-
tation to intensive chemotherapy for acute myeloid leukaemia
in first remission: results of MRC AML 10 trial. Lancet
1998;351:700-708.
56 de Witte T, Van Biezen A, Hermans J et al. Autologous bone
marrow transplantation for patients with myelodysplastic
syndrome (MDS) or acute myeloid leukemia following MDS.
Blood 1997;90:3853-3857.
57 Harousseau JL, Cahn JY, Pignon B et al. Comparison of autol-
ogous bone marrow transplantation and intensive chemother-
apy as postremission therapy in adult acute myeloid leukemia.
Blood 1997;90:2978-2986.
58 Zittoun RA, Mandelli F, Willemze R et al. Autologous or allo-
geneic bone marrow transplantation compared with intensive
chemotherapy in acute myelogenous leukemia. N Engl J Med
1995;332:217-223.
59 Cassileth PA, Harrington DP, Appelbaum FR et al. Chemo-
therapy compared with autologous or allogeneic bone marrow
transplantation in the management of acute myeloid leukemia in
first remission. N Engl J Med 1998;339:1649-1656.
60 Carella AM, Cavaliere M, Lerma E et al. Autologous periph-
eral blood haematopoietic stem cell transplantation for chronic
myelogenous leukaemia. Baillieres Best Pract Res Clin
Haematol 1999;12:209-217.
61 Attal M, Harousseau J-L, Stoppa A-M et al. A prospective,
randomized trial of autologous bone marrow transplantation
and chemotherapy in multiple myeloma. N Engl J Med
1996;335:91-97.
62 Corradini P, Voena C, Astolfi M et al. High-dose sequential
chemoradiotherapy in multiple myeloma: residual tumor cells
are detectable in bone marrow and peripheral blood cell harvests
and after autografting. Blood 1995;85:1596-1602.
63 Lokhorst HM, Schattenberg JJ, Cornelissen JJ et al. Donor
lymphocyte infusions are effective in relapsed multiple
myeloma after allogeneic bone marrow transplantation.
Blood 1997;90:4206-4211.
64 Mehta J, Singhal S. Graft-versus-myeloma. Bone Marrow
Transplant 1998;22:843.
65 Majolino I, Corradini P, Scimè R et al. Allogeneic transplan-
tation of unmanipulated peripheral blood stem cells in
patients with multiple myeloma. Bone Marrow Transplant
1998;22:449-455.
66 Corradini P, Voena C, Tarella C et al. Molecular and clinical
remissions in multiple myeloma: role of autologous and allo-
geneic transplantation of hematopoietic cells. J Clin Oncol
1999;17:208-215.
67 Cavo M, Terragna C, Martinelli G et al. Molecular monitor-
ing of minimal residual disease in patients in long-term com-
plete remission after allogeneic stem cell transplantation for
multiple myeloma. Blood 2000;96:355-357.
68 Nevill TJ, Fung HC, Shepherd JD et al. Cytogenetic abnormal-
ities in primary myelodysplastic syndrome are highly predic-
tive of outcome after allogeneic bone marrow transplantation.
Blood 1998;92:1910-1917.
69 Runde V, de Witte T, Arnold R et al. Bone marrow transplan-
tation from HLA-identical siblings as first-line treatment in
patients with myelodysplastic syndromes: early transplantation
is associated with improved outcome. Bone Marrow Transplant
1998;21:255-261.
70 Anderson JE, Anaseti C, Appelbaum FR et al. Unrelated donor
marrow transplantation for myelodysplasia (MDS) and MDS-
related acute myeloid leukaemia. Br J Haematol 1996;93:59-67.
71 Sutton L, Chastang C, Ribaud P et al. Factors influencing out-
come in de novo myelodysplastic syndromes treated by allo-
geneic bone marrow transplantation: a long-term study of 71
patients. Blood 1996;88:358-365.
by on August 30, 2007 www.StemCells.comDownloaded from
Cutler, Antin
117
72 Appelbaum FR, Anderson J. Allogeneic bone marrow trans-
plantation for myelodysplastic syndrome: outcomes analysis
according to IPSS score. Leukemia 1998;12(suppl 1):S25-S29.
73 Philip T, Guglielmi C, Hagenbeek A et al. Autologous bone
marrow transplantation as compared with salvage chemother-
apy in relapses of chemotherapy-sensitive non-Hodgkins
lymphoma. N Engl J Med 1995;333:1540-1545.
74 Körbling M, Przepiorka D, Huh YO et al. Allogeneic blood
stem cell transplantation for refractory leukemia and lym-
phoma: potential advantage of blood over marrow allografts.
Blood 1995;85:1659-1665.
75 Khouri I, Lee MS, Romaguera J et al. Allogeneic hematopoi-
etic transplantation for mantle-cell lymphoma: molecular
remissions and evidence of graft-versus-malignancy. Ann
Oncol 1999;10:1293-1299.
76 Varadi G, Or R, Kapelushnik J et al. Graft-versus-lymphoma
effect after allogeneic peripheral blood stem cell transplantation
for primary central nervous system lymphoma. Leuk Lymphoma
1999;34:185-190.
77 Corradini P, Ladetto M, Astolfi M et al. Clinical and molecular
remission after allogeneic blood cell transplantation in a patient
with mantle-cell lymphoma. Br J Haematol 1996;94:376-378.
78 Ito M, Shizuru JA. Graft-vs.-lymphoma effect in an allogeneic
hematopoietic stem cell transplantation model. Biol Blood
Marrow Transplant 1999;5:357-368.
79 Storb R, Doney KC, Thomas ED et al. Marrow transplanta-
tion with or without donor buffy coat cells for 65 transfused
aplastic anemia patients. Blood 1982;59:236-246.
80 Anasetti C, Storb R, Longton G et al. Donor buffy coat cell
infusion after marrow transplantation for aplastic anemia
[Letter]. Blood 1988;72:1099-1100.
81 Flowers MED, Leisenring W, Beach K et al. Granulocyte
colony-stimulating factor given to donors before apheresis
does not prevent aplasia in patients treated with donor leuko-
cyte infusion for recurrent chronic myeloid leukemia after
bone marrow transplantation. Biol Blood Marrow Transplant
2000;6:321-326.
82 Porter DL, Roth MS, McGarigle C et al. Induction of graft-
versus-host disease as immunotherapy for relapsed chronic
myeloid leukemia. N Engl J Med 1994;330:100-106.
83 Khouri IF, Keating M, Körbling M et al. Transplant-lite: induc-
tion of graft-versus-malignancy using fludarabine-based non-
ablative chemotherapy and allogeneic blood progenitor-cell
transplantation as treatment for lymphoid malignancies. J Clin
Oncol 1998;16:2817-2824.
84 Nagler A, Slavin S, Varadi G et al. Allogeneic peripheral
blood stem cell transplantation using a fludarabine-based low
intensity conditioning regimen for malignant lymphoma.
Bone Marrow Transplant 2000;25:1021-1028.
by on August 30, 2007 www.StemCells.comDownloaded from
DOI: 10.1634/stemcells.19-2-108
2001;19;108-117 Stem Cells
Corey Cutler and Joseph H. Antin Peripheral Blood Stem Cells for Allogeneic Transplantation: A Review
This information is current as of August 30, 2007
& Services
Updated Information http://www.StemCells.com/cgi/content/full/19/2/108
including high-resolution figures, can be found at:
by on August 30, 2007 www.StemCells.comDownloaded from
... Stem cell transplant is performed in settings that damage the body's stem cells, including hematologic malignancies (e.g., leukemia, lymphoma, multiple myeloma, neuroblastoma) or cancer therapy (e.g., high-dose chemotherapy, total body irradiation) (192). Stem cell transplant relies on 3 stem cell sources: bone marrow, peripheral blood, and umbilical cord blood (193). ...
... PBSCs are collected by continuous-flow apheresis after mobilization using medications including granulocyte colony-stimulating factor (G-CSF) agents and chemokine receptor 4 (CXCR4) blockers (e.g., plerixafor); chemotherapy can also be used for mobilization (i.e., chemoembolization) (200). PBSCs are generally identified and quantified using flowcytometry via their immunophenotypic patterns (e.g., CD34 + CD38 − ) (192). Besides CD34 + HSC subpopulations, PBSC grafts contain nucleated cells including DCs, T cells, B cells, NK cells, and monocytes (201). ...
Article
Full-text available
Cell therapy practices date back to the 19 th century and continue to expand on investigational and investment grounds. Cell therapy includes stem cell- and non–stem cell-based, unicellular and multicellular therapies, with different immunophenotypic profiles, isolation techniques, mechanisms of action, and regulatory levels. Following the steps of their predecessor cell therapies that have become established or commercialized, investigational and premarket approval-exempt cell therapies continue to provide patients with promising therapeutic benefits in different disease areas. In this review article, we delineate the vast types of cell therapy, including stem cell-based and non–stem cell-based cell therapies, and create the first-in-literature compilation of the different “multicellular” therapies used in clinical settings. Besides providing the nuts and bolts of FDA policies regulating their use, we discuss the benefits of cell therapies reported in 3 therapeutic areas—regenerative medicine, immune diseases, and cancer. Finally, we contemplate the recent attention shift toward combined therapy approaches, highlighting the factors that render multicellular therapies a more attractive option than their unicellular counterparts.
... After priming, peripheral venous apheresis by extracorporeal continuous-flow machines processes 2-3 units of patient blood, segregating stem cells and granulocytes and returning red blood cells and plasma fractions to the patient. Generally, after 1-2 sessions lasting 3-5 hours, sufficient levels of PBSCs are collected [25]. Further isolation of PBSCs is necessary to limit the risk of GvHD due to potential contamination with donor T-cells [26]. ...
Article
Mesenchymal stem cells hold many therapeutic benefits in treating diverse disease states, but autologous requirements, high costs, lack of standardization and other factors limit their widespread application. Additionally, researchers have discovered that many mesenchymal stem cell in-vivo benefits originate from their paracrine factors. Therefore, cell-free therapeutics, including mesenchymal stem cell-derived conditioned medium and extracellular vesicles have been suggested as alternative bioregenerative therapies. This literature review summarizes mesenchymal stem cell application, the benefits of cell-free strategies and the components of its secretome. This manuscript is a companion piece to “Part 2: Bioregenerative Applications of the Human Mesenchymal Stem Cell-Derived Secretome,” included in this issue, which contains the results of in-vivo studies applying the conditioned medium and extracellular vesicles to human and animal models, ongoing clinical trials, limitations to cell-free strategies and future directions for the wide-scale adoption of these therapies.
... Both autologous and allogeneic PBSCT can be used depending on the individual patient's condition and treatment objectives. Studies of the outcomes of PBSCT have been conducted based on the number of infused CD34 + cells, and 2-5 × 10 6 cells/kg of the recipient's body weight has been suggested as the optimal collection amount for a successful transplantation outcome [1][2][3]. Because the collection of sufficient CD34 + cells is important for successful transplantation, CD34 + cell mobilization using G-CSF and chemotherapy prior to PBSCH is a pivotal step. ...
Article
Full-text available
Simple Summary This study aims to describe the factors associated with poor mobilization in patients undergoing autologous peripheral blood stem cell harvest. Poor mobilization was observed in 30.5% of the patients included in the investigation. While older age, myelotoxic agents, and low platelet counts were associated with poor mobilization in patients with hematologic diseases, a history of radiation therapy to the spine and pelvic region was associated with mobilization failure in patients with non-hematologic diseases. In patients who received plerixafor, 14.2% suffered from mobilization failure, with differences in hemoglobin, platelet counts, and mobilization regimens noted between the poor and good mobilization groups. Identifying potential risk factors for poor mobilization in specific groups of patients will aid in planning therapy and ensuring successful transplantation in those requiring hematopoietic stem cell transplants. Abstract Peripheral blood stem cell transplantation (PBSCT) is an important therapeutic measure for both hematologic and non-hematologic diseases. For PBSCT to be successful, sufficient CD34⁺ cells need to be mobilized and harvested. Although risk factors associated with poor mobilization in patients with hematologic diseases have been reported, studies of patients with non-hematologic diseases and those receiving plerixafor are rare. To identify factors associated with poor mobilization, data from autologous PBSC harvest (PBSCH) in 491 patients were retrospectively collected and analyzed. A multivariate analysis revealed that in patients with a hematologic disease, an age older than 60 years (odds ratio [OR] 1.655, 95% confidence interval [CI] 1.049–2.611, p = 0.008), the use of myelotoxic agents (OR 4.384, 95% CI 2.681–7.168, p < 0.001), and a low platelet count (OR 2.106, 95% CI 1.205–3.682, p = 0.009) were associated with poor mobilization. In patients with non-hematologic diseases, a history of radiation on the pelvis/spine was the sole associated factor (OR 12.200, 95% CI 1.934–76.956, p = 0.008). Among the group of patients who received plerixafor, poor mobilization was observed in 19 patients (19/134, 14.2%) and a difference in the mobilization regimen was noted among the good mobilization group. These results show that the risk factors for poor mobilization in patients with non-hematologic diseases and those receiving plerixafor differ from those in patients with hematologic diseases; as such, non-hematologic patients require special consideration to enable successful PBSCH.
... As células do sangue periférico ganharam uma importante utilização nos últimos anos em decorrência da sua aplicação para transplante de células hematopoiéticas em substituição ao transplante de medula óssea, e são representadas por subpopulação de células CD34+ após a aplicação de fatores que estimulam colônias (CUTLER;ANTIN, 2001).Estudos recentes mostraram que mesmo em indivíduos adultos, a neurogénese, ou seja, renovação celular do sistema nervoso através de formação, migração e diferenciação de novos neurônios, ocorre em diversas regiões do cérebro, que possuem células-tronco neurais adultas multipotentes como componentes de astrócitos e células ependimais nas zonas subventriculares adjacentes (GRITTI; VESCOVI; GALLI, 2002). Essas células possuem a capacidade de originarem células sanguíneas, astrócitos, neurônios e oligodendrócitos, e são estimuladas a proliferação quando em resposta a fatores que induzem a mitose, como por exemplo o fator de crescimento epidérmico (EGF) e o fator de crescimento de fibroblasto 2 (FGF-2) (VILLA et al., 2001). ...
Article
Full-text available
O corpo humano é composto por centenas de células, dentre essas centenas, existem as denominadas células tronco, que possuem a capacidade de dar origem a diversos tecidos e são responsáveis por formarem nossos órgãos. É possível que através desse potencial regenerador das células tronco sobre as células nervosas, as células tronco desempenhem um efeito terapêutico sobre as neuropatias, que são consequências de disfunções ou lesões no sistema nervoso. Estudos recentes corroboram que a administração de células tronco pode levar à redução de dores neuropáticas comportamentais não só em modelos experimentais com ligadura de nervo isquiático, mas também com a neuropatia diabética. O presente estudo tem como objetivo principal revisar a literatura sobre quais as formas e quais as utilidades de células-tronco para tratamento de dores neuropáticas. O método utilizado para a realização deste estudo foi a revisão bibliográfica sistemática, e os resultados foram obtidos de oito publicações selecionadas. Concluiu-se com esse trabalho que existem diversos registros que corroboram os efeitos positivos obtidos no tratamento para dor neuropática utilizando células-tronco transplantadas de diferentes origens e para diferentes tratamentos de dores, mas ressalta-se que mais pesquisas devem ser feitas sobre o assunto para padronização do tratamento.
... These values are similar to those generally expected when HSCT is performed using peripheral blood. 9 In addition, only 9 (7.8%) patients experienced primary graft failure (data not shown), which is comparable with the result of Basak, et al. 4 (7.8% vs. 6.0%). All patients who failed engraftment received peripheral blood stem cells infusion. ...
Article
Full-text available
Purpose: The purpose of this study is to share our outcomes and experiences on allogeneic hematopoietic stem cell transplantation (HSCT) in elderly patients aged 60 years and older with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) in South Korea, and to compare them with other studies. Materials and methods: We analyzed the clinical outcomes of 116 patients with AML or MDS aged 60 years and older who underwent allogeneic HSCT. We also analyzed which pretreatment factors affect the overall survival (OS) after allogeneic HSCT. Results: Neutrophil and platelet engraftment were achieved at median day +11 [interquartile range (IQR) 10-15] and +14 (IQR 11-19), respectively. A complete donor chimerism was confirmed in 65 (56.0%) patients at 3 weeks and in 63 (54.3%) patients at 3 months after HSCT. The estimated incidence of grade II-IV acute graft-versus-host disease (GVHD) at day 100 was 13.7%. The estimated incidence of chronic GVHD at 2 years was 38.8%. Within a median follow-up of 14 months after HSCT, OS was 64% at 1 year and 55% at 2 years, and non-relapse mortality (NRM) was 20% at 1 year and 28% at 2 years. Multivariate analysis revealed that male sex and Hematopoietic Cell Transplantation-Specific Comorbidity Index ≥3 were associated with poor OS. Conclusion: This study showed that allogeneic HSCT in elderly adults aged 60 and older can be performed with successful engraftment and acceptable NRM and OS are expected given the generally known survival of patients with higher risk MDS and poor risk AML.
... In medicine, cell and gene therapy are founded on the ability to transplant stem cells from a healthy donor or after corrective gene manipulation to a sick recipient. The integration and activity of the self-perpetuating population of healthy cells are the basis for the patient's therapy [8][9][10][11][12][13] . Historically, stem cell manipulation and transplantation formed the foundation for understanding the development and maintenance of tissue and organ function, especially the hematopoietic and immune systems 9,12,13 . ...
Preprint
Full-text available
Stem cells are the base for cell therapy due to their ability to self-renew, differentiate into other cell types, and live throughout the life of an organism. The initial cell therapy was bone marrow transplantation; bone marrow that contains haematopoietic stem cells is transferred from a healthy donor into a sick recipient to transfer the healthy genotype 1,2 . Stem cell therapy approach may be possible in corals, where there is genotypic variation in heat tolerance that influences survival during increased water temperatures 3–5 . However, we first need the ability to reliably isolate and transplant stem cells in Hexacorallia, which includes stony corals and sea anemones ⁶ . In this work, we used the sea anemone Nematostella vectensis as our model for candidate stem cell transplantation, as it is the only hexacorallian species that has fluorescent-tagged transgenic strains ⁷ . We established cell transplantation to show that there are cell populations exhibiting the functional characteristics of stem cells. We showed that a subpopulation of cells from N. vectensis can be transplanted from donor to recipient, are long-lived and self-renewing, can proliferate and differentiate, can integrate into the recipient, and rescue recipient animals treated with lethal doses of a chemotherapeutic agent. Lastly, we showed that this subpopulation can be enriched by sorting, using species non-specific cell markers and that similar subpopulations of cells can be isolated from other hexacorallians, including stony corals. This lays a foundation for the possibility of stem cell-based therapy in species of Hexacorallia.
... As technology and methods improved however, it was found that treating patients with a cytokine treatment prior to peripheral blood progenitor cell harvesting mobilized many of the desired cells into the blood stream and drastically increased the efficacy of this technique, making it clinically viable. [31][32][33] In a double blinded randomized study 40 patients underwent bone marrow and peripheral blood progenitor cell collections and the yield of useable harvested cells were compared. It was found that blood progenitor cell collection yielded significantly more useable stem cells and patients were able to undergo the collection procedure more frequently when compared to the bone marrow harvesting method. ...
Article
Stem cells are types of cells that have unique ability to self-renew and to differentiate into more than one cell lineage. They are considered building blocks of tissues and organs. Over recent decades, they have been studied and utilized for repair and regenerative medicine. One way to classify these cells is based on their differentiation capacity. Totipotent stem cells can give rise to any cell of an embryo but also to extra-embryonic tissue as well. Pluripotent stem cells are limited to any of the three embryonic germ layers; however, they cannot differentiate into extra-embryonic tissue. Multipotent stem cells can only differentiate into one germ line tissue. Oligopotent and unipotent stem cells are seen in adult organ tissues that have committed to a cell lineage. Another way to differentiate these cells is based on their origins. Stem cells can be extracted from different sources, including bone marrow, amniotic cells, adipose tissue, umbilical cord, and placental tissue. Stem cells began their role in modern regenerative medicine in the 1950’s with the first bone marrow transplantation occurring in 1956. Stem cell therapies are at present indicated for a range of clinical conditions beyond traditional origins to treat genetic blood diseases and have seen substantial success. In this regard, emerging use for stem cells is their potential to treat pain states and neurodegenerative diseases such as Parkinson’s and Alzheimer’s disease. Stem cells offer hope in neurodegeneration to replace neurons damaged during certain disease states. This review compares stem cells arising from these different sources of origin and include clinical roles for stem cells in modern medical practice.
... Currently, bone marrow (BM), peripheral blood (PB) and umbilical cord blood (UCB) serve as cell sources for HSC transplantation. Among these, in the last decades UCB has attracted increasing attention because UCB have several advantages: their collection is non-invasive and safe for the donor, permissive of HLA mismatch and lower risk of graft-versus-host disease compared to BM or PB-derived CD34+ hematopoietic stem and progenitor cells (HSPCs) [2,3]. Nonetheless, utilization of UCB is limited as doses of HSPCs are lower in the UCB graft, which associated with inefficient engraftment and treatment-related mortality [4,5]. ...
Article
Full-text available
Hematopoietic stem and progenitor cell (HSPC) transplantation is a curative treatment of hematological disorders that has been utilized for several decades. Although umbilical cord blood (UCB) is a promising source of HSPCs, the low dose of HSPCs in these preparations limits their use, prompting need for ex vivo HSPC expansion. To establish a more efficient method to expand UCB HSPCs, we developed the bioactive peptide named SL-13R and cultured UCB HSPCs (CD34+ cells) with SL-13R in animal component-free medium containing a cytokine cocktail. Following 9 days of culture with SL-13R, the numbers of total cells, CD34+, CD38− cells, and hematopoietic stem cell (HSC)-enriched cells were significantly increased relative to control. Transplantation of cells cultured with SL-13R into immunodeficient NOD/Shi-scid/IL-2Rγ knockout mice confirmed that they possess long-term reconstitution and self-renewal ability. AHNAK, ANXA2, and PLEC all interact with SL-13R. Knockdown of these genes in UCB CD34+ cells resulted in reduced numbers of hematopoietic colonies relative to SL-13R-treated and non-knockdown controls. In summary, we have identified a novel bioactive peptide SL-13R promoting expansion of UCB CD34+ cells with long-term reconstitution and self-renewal ability, suggesting its clinical use in the future.
Article
Full-text available
Hematopoietic stem cell transplantation is a therapeutic procedure in which the hematopoietic stem cells, derived from any source and from any donor, are administered to the recipient, in order to replace the hematopoiesis, completely or partially. Due to clinical experience and progress made in the last five decades, hematopoietic stem cell transplantation is used as a standard procedure for treating a variety of malignant, hematologic, immunologic, metabolic, autoimmune and other serious diseases. In late 1980s and early 1990s, it was discovered that human umbilical cord blood is a rich source of hematopoietic stem cells. In the beginning, hematopoietic stem cell transplantations were performed only in children. It has been observed that the graft-versus-host disease is rarer and less conspicuous after hematopoietic stem cell transplantation obtained from umbilical cord blood, compared with transplantation obtained from the bone marrow and peripheral blood, even in the presence of human leukocyte antigen mismatch (HLA-mismatch). It was concluded that the umbilical cord blood can be used as an alternative source of hematopoietic stem cells for transplantation from unrelated donors. The organization of umbilical cord blood banks is essential, in order to establish widespread clinical use of this type of transplantation. The first umbilical cord blood banks are formed in the early 1990s in New York, Milan and Dusseldorf, and during the 1990s, numerous banks are formed in the USA, Europe, China, Japan and Australia. At present, there are 158 public banks with more than 700.000 cryopreserved umbilical cord blood samples.
Chapter
Stem cells have the remarkable capability to self-renew in an undifferentiated state and to differentiate into many types of cells with specific functions upon receiving appropriate triggers. In this article, five different types of stem cells are discussed: human embryonic stem cells, human-induced pluripotent stem cells, neural stem cells, mesenchymal stem cells, and hematopoietic stem cells. We describe their initial discovery, sources where they are obtained and niches where these stem cells reside, the canonical phenotypic markers for distinguishing them, their lineage differentiation capabilities, and review some key clinical trials that are ongoing for each of these important stem cell types.
Article
Full-text available
Sixty-five multiply transfused patients with severe aplastic anemia were given cyclophosphamide followed by grafts anemia were given cyclophosphamide followed by grafts from HLA-identical siblings. The effect of the administration of viable donor buffy coat cells following the marrow inoculum was evaluated with regard to graft rejection and survival. Results in 43 patients so treated are presented along with those in 22 concurrent patients given marrow alone. Most patients given buffy coat had positive in vitro tests of sensitization indicating a high risk for graft rejection, while all but one of the patients given marrow alone had negative tests. Thirty of the 43 (70%) patients given marrow and buffy coat are alive between 10 and 61 mo (median 36) after grafting; 4 died after graft rejection and 6 with acute or chronic graft-versus-host disease (GVHD). Eleven of the 22 (50%) patients given marrow alone are alive between 29 and 65 mo (median 52); 7 died after graft rejection and 3 with GVHD. The addition of buffy coat cell infusions to the marrow inoculum reduced the risk of rejection and increased survival in the currently reported transfused patients when compared to patients grafted before 1976. However, there was an increased risk of chronic GVHD. Recipients of marrow from female donors survived slightly better (73%) than recipients of male marrow (58%).
Article
The detection of residual molecular and cytogenetic disease was prospectively compared in patients with Philadelphia-chromosome (Ph1) positive first chronic phase chronic myelogenous leukemia (CML) who underwent allogeneic transplantation of unmanipulated peripheral blood stem cells (PBSCT) (n = 29) or bone marrow (BM) (n = 62) using genotypically HLA-identical sibling donors or partially HLA-matched extended family donors. A molecular relapse (MR), as defined by two consecutive positive polymerase chain reaction (PCR) assays for the detection of M-bcr-abl transcripts in a 4-week interval, was found in two of 29 (7%) patients after PBSCT compared with 20 of 62 (32%) patients after bone marrow transplantation (BMT). This corresponds to a 4-year molecular relapse estimate (± standard error) of 7% ± 5% after PBSCT and of 44% ± 8% after BMT (P < .009). With identical follow-up periods of survivors in both patient subsets between 6 and 55 months (median, 28 months), 14 of the 20 patients with MR after BMT progressed to an isolated cytogenetic (n = 10) or a hematologic (n = 4) disease recurrence, resulting in a 4-year cytogenetic relapse estimate of 47% ± 11%, while none of the patients after PBSCT has so far relapsed (P < .006). Multivariate analysis including all potential influencial factors of posttransplant disease recurrence identified the source of stem cells (P < .02) as the only independent predictor of molecular relapse. In conclusion, this prospective comparison of molecular and cytogenetic residual disease demonstrates that peripheral blood stem cell transplants have a more pronounced activity against residual CML cells than bone marrow transplants. Prospective randomized trials comparing PBSCT and BMT in patients with first chronic phase Ph1-positive CML are strictly required to further substantiate differences in the antileukemic activity of the two stem cell sources.
Article
We evaluated demographic characteristics and graft composition as risk factors for acute graft-versus-host disease (GVHD) in 160 adult recipients of HLA-identical allogeneic blood stem cell transplants. The patients received a median nucleated cell dose of 7.9 × 108/kg and median C34+ cell dose of 5.6 × 106/kg. GVHD prophylaxis consisted of cyclosporine (CSA) and steroids, tacrolimus (FK506) and steroids, or FK506 and methotrexate. Grades 2 to 4 GVHD occurred in 31% (95% CI, 23% to 39%), and grades 3 to 4 GVHD in 14% (95% CI, 8% to 20%). In univariate analyses, GVHD prophylaxis with CSA and high CD34+ cell doses were significant risk factors for grades 2 to 4 GVHD, but diagnosis, age, use of total body irradiation, donor sex, female donor for male recipient, donor parity, donor alloimmunization, viral serology, nucleated cell dose, CD3+ cell dose, and CD56+ cell dose did not alter the incidence of GVHD significantly. With a CD34+cell dose less than 8 × 106 CD34+ cells/kg, the risk of grades 2 to 4 GVHD was significantly higher for those who received CSA (39%, 95% CI, 21% to 47%) in comparison with those on FK506 (18%, 95% CI, 10% to 26%) (P = .03), but GVHD prophylaxis regimen had less impact with a higher CD34+cell dose (overall grades 2 to 4 GVHD rate 52%, 95% CI, 37% to 67%). GVHD prophylaxis and CD34+ cell dose are independent risk factors for acute GVHD after allogeneic blood stem cell transplantation.
Article
In the present study, we used a polymerase chain reaction–based (PCR-based) strategy to retrospectively analyze the presence of residual myeloma cells in serial posttransplant bone marrow samples obtained from 13 patients in remission after allogeneic hemopoietic stem cell transplantation (allo SCT). For this purpose, patient-specific primers were generated from complementarity determining regions 2 and 3 of the rearranged IgH gene. The level of sensitivity of the PCR-based assay ranged from 1 in 105 to 1 in 106 normal marrow cells. Following transplantation, 9 of 12 patients who attained stringently defined complete remission (CR) remained persistently PCR− for a median of 36 months, and 4 of the patients remained PCR− up to the latest analysis, which was performed at 48, 72, 72, and 120 months, respectively, after allo SCT. None of the patients in the PCR− subgroup experienced a disease relapse, and only 1 of 4 PCR+ patients experienced a relapse. It is concluded that allo SCT has the potential ability to induce sustained serological and molecular CR in selected patients with multiple myeloma.
Article
Allogeneic bone marrow transplantation (BMT) is the only curative therapy available for patients with myelodysplastic syndrome (MDS). In an attempt to identify prognostic factors influencing outcome, we collected data retrospectively on 60 consecutive adult patients who had undergone BMT at our center for primary MDS or acute myelogenous leukemia evolving from preexisting primary MDS (sAML). Patients were divided into subgroups according to cytogenetic abnormalities based on a recently described International MDS Workshop categorization system. The 7-year actuarial event-free survival (EFS), relapse rate, and nonrelapse mortality (NRM) for all patients were 29% (95% confidence interval [CI], 16% to 43%), 42% (CI, 24% to 67%), and 50% (CI, 37% to 64%), respectively. The EFS for the good-, intermediate-, and poor-risk cytogenetic subgroups were 51% (CI, 30% to 69%), 40% (CI, 16% to 63%), and 6% (CI, 0% to 24%), respectively (P= .003). The corresponding actuarial relapse rates were 19% (CI, 6% to 49%), 12% (CI, 2% to 61%), and 82% (CI, 48% to 99%), respectively (P = .002) with no difference in NRM between the subgroups. Univariate analysis showed cytogenetic category, French-American-British (FAB) subtype, and graft-versus-host disease (GVHD) prophylaxis used to be predictive of relapse and EFS. In multivariate analysis, only the cytogenetic category was predictive of EFS, with the relative risk of treatment failure for the good-, intermediate-, and poor-risk cytogenetic subgroups being 1.0, 1.5, and 3.5, respectively (P = .004). For adults with primary MDS and sAML, even after BMT, poor-risk cytogenetics are predictive of an unfavorable outcome; novel treatment strategies will be required to improve results with allogeneic BMT in this patient population. © 1998 by The American Society of Hematology.
Article
We report on 71 consecutive patients with de novo myelodysplastic syndromes referred to physicians belonging to the Societe francaise de greffe de moelle from 1982 through 1991 and transplanted with marrow from HLA-identical siblings. There were 16 cases of refractory anemia, 27 of refractory anemia with excess of blast cells, and 28 of refractory anemia with excess of blast cells in transformation. Seventeen patients had received cytoreductive chemotherapy before the graft. The disease progressed in 17 patients between diagnosis and grafting. Twenty-three patients are alive with a median follow-up of 6 years, whereas 24 died from relapse and 24 from transplant-related complications. Kaplan-Meier estimates of event-free survival, relapse and transplant-related mortality at 7 years were 32%, 48%, and 39%, respectively. The log-rank test and Cox's model revealed better outcome among young patients, patients in an early stage of the French-American- British (FAB) classification or with a low percentage of marrow blasts before transplantation, patients who did not undergo cytoreductive chemotherapy before transplantation, and patients conditioned with total body irradiation and cyclophosphamide. The high rate of relapse in advanced FAB stages has led us to graft patients earlier in the course of the disease, and we are currently conducting a multicenter, randomized study to determine the value of intensive chemotherapy before grafting in patients with an excess of marrow blasts.
Article
Peripheral blood stem cells (PBSC) obtained from granulocyte-colony stimulating factor (G-CSF)-mobilized donors are increasingly used for allogeneic transplantation. Despite a 10-fold higher dose of transplanted T cells, acute graft-versus-host disease (GVHD) does not develop in higher proportion in recipients of PBSC than in recipients of marrow. T cells from G-CSF-treated experimental animals preferentially produce IL-4 and IL-10, cytokines characteristic of Th2 responses, which are associated with diminished GVHD-inducing ability. We hypothesized that G-CSF-mobilized PBSC contain antigen-presenting cells, which prime T-lymphocytes to produce Th2 cytokines. Two distinct lineages of dendritic cells (DC) have been described in humans, DC1 and DC2, according to their ability to induce naive T-cell differentiation to Th1 and Th2 effector cells, respectively. We have used multicolor microfluorometry to enumerate DC1 and DC2 in the peripheral blood of normal donors. G-CSF treatment with 10 to 16 μg/kg per day for 5 days increased peripheral blood DC2 counts from a median of 4.9 × 106/L to 24.8 × 106/L (P = .0009), whereas DC1 counts did not change. Purified DC1, from either untreated or G-CSF treated donors, induced the proliferation of allogeneic naive T cells, but fresh DC2 were poor stimulators. Tumor necrosis factor- (TNF-)-activated DC1 induced allogeneic naive T cells to produce IFN-γ, which is typical of Th1 responses, whereas TNF--activated DC2 induced allogeneic naive T cells to produce IL-4 and IL-10, which are typical of Th2 responses. PBSC transplants contained higher doses of DC2 than marrow transplants (median, 2.4 × 106/kg versus 0.5 × 106/kg) (P = .006), whereas the dose of DC1 was comparable. Thus, it is conceivable that transplantation of G-CSF-stimulated PBSC does not result in overwhelming acute GVHD because the graft contains predominantly Th2-inducing DC. Adoptive transfer of purified DC2 may be exploited to induce immune deviation after transplantation of hematopoietic stem cells or organ allografts.
Article
Chronic graft-versus-host disease (GVHD) is likely caused by donor T lymphocytes. Because unmodified blood stem cell grafts contain one log more T lymphocytes than unmodified marrow grafts, we evaluated the incidence of chronic GVHD in previously reported 37 blood stem cell recipients and 37 computer-matched historical control marrow recipients (Bensinger et al, Blood 88:2794, 1996). All patients have been followed until death, relapse, or occurrence of chronic GVHD or for a minimum of 2 years. In a univariable proportional hazards regression model, the relative risk of developing clinical chronic GVHD (includes clinical limited and clinical extensive disease) by 2 years posttransplant among the peripheral blood stem cell recipients compared with the marrow recipients was 2.22 (95% confidence interval, 1.04 to 4.74; P = .039). For clinical extensive chronic GVHD, the relative risk was 2.37 (95% confidence interval, 1.07 to 5.29; P = .035). In multivariable analyses, considering also the covariables of patient age, patient cytomegalovirus serostatus, and donor cytomegalovirus serostatus, the relative risks of clinical chronic GVHD and clinical extensive chronic GVHD were also greater than 2 (P < .05). We conclude that the transplantation of unmanipulated filgrastim-mobilized blood stem cells may result in a relatively high incidence of chronic GVHD.
Article
The effects of daily administration of recombinant human granulocyte colony-stimulating factor (rhG-CSF) to eight normal volunteers donating granulocytes for neutropenic relatives undergoing marrow transplantation were studied. Granulocyte donors consisted of seven marrow donors (5 syngeneic, 2 HLA identical) and one haploidentical son who had not donated marrow. All donors were administered daily rhG-CSF at a mean dose of 5 micrograms/kg/d (range 3.5 to 6.0) for a mean of 11.75 days (range 9 to 14 days), and granulocytes were collected a mean of 7.6 times (range 4 to 12). RhG-CSF was well tolerated and only minor side effects were observed. All donors became anemic from marrow donation and the removal of red blood cells during the collection procedures. Red blood cell transfusions were not given. All donors had a decrease in platelet counts and the magnitude of the decrement appeared to be greater than in historical donors. This was due in part to increased removal of platelets with the collection product, but a direct effect of rhG-CSF on platelet production cannot be excluded. The mean precollection granulocyte level was 29.6 x 10(9)/L (range 11.8 to 79.8), which was a 10-fold increase over baseline. The mean number of granulocytes collected was 41.6 x 10(9) (range 1.3 to 144.1), which was a six-fold increase over historical donors not receiving rhG-CSF. The mean granulocyte level 24 hours after transfusion into neutropenic recipients was 0.95 x 10(9)/L (median 0.57 and range .06 to 9.47). This study indicates that rhG-CSF is safe to administer to normal individuals, significantly improves the quantity of granulocytes collected, and results in significant circulating levels of granulocytes in neutropenic recipients. Further studies to evaluate rhG- CSF in normal granulocyte donors are warranted.
Article
We suggest that acute GVHD after marrow transplantation reflects (1) host injury due to the conditioning regimen followed by the production of inflammatory cytokines; (2) stimulation of mature donor T cells in the milieu of increased cell surface expression of leukocyte adhesion molecules and HLA molecules, followed by the autocrine production of IL- 2; and, finally, (3) recruitment and activation of additional mononuclear effector cells from donor marrow progenitors, which produce additional inflammatory cytokines, thus sustaining the response. The second step is critical for the amplification of the systemic inflammatory response, and it is absence in autologous, syngeneic, and T-cell-depleted transplants. These T cells may also contribute to the inflammatory cytokine network. Acute GVHD can occur in the absence of primary tissue injury in such settings as transfusion-related GVHD; however, it is likely that a greater HLA disparity between donor and host is required. We propose that inflammatory cytokine production is the final common pathway of acute GVHD. If this model is correct, control of cytokine dysregulation at any of several points should control GVHD. Further studies of GVHD and investigations of cytokine antagonists (eg, IL-4 or IL-10) or combinations of antagonists such as IL-1ra and soluble TNF receptor or pentoxifylline will allow us to determine the validity of this hypothesis.