ArticlePDF Available

Variable product purity and functional capacity after CD34 selection: a direct comparison of the CliniMACSR (v2.1) and IsolexR 300i (v2.5) clinical scale devices

Wiley
British Journal of Haematology
Authors:

Abstract and Figures

The two clinical scale devices currently available for CD34+ cell selection from peripheral blood stem cells (PBSC) apheresis products, the CliniMACS and the Isolex 300i, were compared directly by pooling and splitting two PBSC harvests collected on sequential days from 10 patients and processing half of each pooled harvest on each device. The CliniMACS product had significantly higher median CD34+ purity (90%vs 78%; P = 0.004) and lower median T-cell content (0.06%vs 0.44%; P = 0.003) compared with the Isolex 300i product. The median CD34+ yields were similar (64% and 60% respectively). However, when the functional capacities of the products were compared, the median recovery of colony-forming units was significantly greater from the Isolex 300i product (48%vs 38%; P = 0.035), as was expansion of cells in either erythroid or granulocytic lineage-specific liquid culture (2.1-fold more erythroid and 1.5-fold more granulocytic lineage progenitors on d 9 (P = 0.03 and 0.03 respectively). This was due to a higher proportion of apoptotic cells in the CliniMACS product (28%vs 18%; P = 0.007, annexin V binding). Hence, although the CliniMACS device yielded a higher purity product with fewer T cells, the Isolex 300i product contained fewer apoptotic cells and consequently had greater functional capacity in culture.
Content may be subject to copyright.
Variable product purity and functional capacity after CD34
selection: a direct comparison of the CliniMACS
(v2Æ1)
and Isolex
300i (v2Æ5) clinical scale devices
Michael J. Watts, Tim C. P. Somervaille, Stuart J. Ings, Forhad Ahmed, Asim Khwaja, Kwee Yong
and David C. Linch Department of Haematology, Royal Free and University College London Medical School, London,
UK
Received 8 October 2001; accepted for publication 12 December 2001
Summary. The two clinical scale devices currently avail-
able for CD34
+
cell selection from peripheral blood stem
cells (PBSC) apheresis products, the CliniMACS and the
Isolex 300i, were compared directly by pooling and split-
ting two PBSC harvests collected on sequential days from
10 patients and processing half of each pooled harvest on
each device. The CliniMACS product had significantly
higher median CD34
+
purity (90% vs 78%; P¼0Æ004)
and lower median T-cell content (0Æ06% vs 0Æ44%;
P¼0Æ003) compared with the Isolex 300i product. The
median CD34
+
yields were similar (64% and 60%
respectively). However, when the functional capacities of
the products were compared, the median recovery of col-
ony-forming units was significantly greater from the Isolex
300i product (48% vs 38%; P¼0Æ035), as was expansion
of cells in either erythroid or granulocytic lineage-specific
liquid culture (2Æ1-fold more erythroid and 1Æ5-fold more
granulocytic lineage progenitors on d 9 (P¼0Æ03 and
0Æ03 respectively). This was due to a higher proportion of
apoptotic cells in the CliniMACS product (28% vs 18%;
P¼0Æ007, annexin V binding). Hence, although the
CliniMACS device yielded a higher purity product with
fewer T cells, the Isolex 300i product contained fewer
apoptotic cells and consequently had greater functional
capacity in culture.
Keywords: CliniMACS
, (v2Æ1), Isolex
, 300i (v2Æ5), CD34
+
selection, CD34
+
product functional capacity, apoptosis.
Peripheral blood stem cells (PBSCs), mobilized with a variety
of growth factor and chemotherapeutic regimens, are now
used extensively in autologous and allogeneic transplanta-
tion (Schmitz et al, 1996; Gratwohl et al, 2001). Many
PBSC harvests are used directly without ex vivo manipula-
tion. However, where there is a requirement either for T-cell
depletion of the harvest to reduce the risk of graft-versus-
host disease following haploidentical and other allogeneic
transplants (Urbano-Ispizua et al, 1997; Kawano et al,
1998), or for purging of malignant cells from the harvest
(Cagnoni & Shpall, 1996), selection of CD34
+
progenitor
cells before transplantation may be carried out. Addition-
ally, CD34
+
cell selection is a prerequisite for some gene
therapy procedures (Devereux et al, 1998), as well as other
experimental approaches involving clinical scale ex vivo
expansion of haemopoietic cells for transplantation (Reiffers
et al, 1999; Ardeshna et al, 2000).
Two devices are currently available for clinical scale
CD34
+
cell selection from PBSC apheresis products: the
CliniMACS (Miltenyi Biotec GmbH, Bergisch Gladbach,
Germany) and the Isolex 300i (Baxter Healthcare, Deerfield,
IL). These devices differ in the manner in which the CD34
+
cells are isolated. The CliniMACS device utilizes a monoclo-
nal anti-CD34 antibody (QBEnd10), directly conjugated to
an iron oxide/dextran particle approximately 50 nm in size.
The conjugated antibody binds CD34
+
cells and these are
captured using high-gradient magnetic fields created by pla-
cing a column of small ferromagnetic beads between the poles
of rare earth magnets. To enhance the purity of the CD34
cells, three rounds of magnetic selection are used. The CD34
+
cells in the resultant product retain the ferromagnetic
particles following selection. The Isolex 300i utilizes a
different murine monoclonal anti-CD34 antibody (9C5),
which is unconjugated. Following coating of the CD34
+
cells in the harvest with the antibody, cells are rosetted with
magnetic beads (4Æ5lm in size) coated with polyclonal
Correspondence: Michael J. Watts, Department of Haematology,
Royal Free and University College Medical School, Department of
Haematology, Royal Free and University College London Medical
School, 98 Chenies Mews, London WC1E 6HX, UK. E-mail:
michael.watts@ucl.ac.uk
British Journal of Haematology, 2002, 118, 117–123
2002 Blackwell Science Ltd 117
sheep anti-mouse immunoglobulin (Dynabeads). Addition-
ally, DNAase may be added to reduce cell clumping and
increase yield and purity (Fritsch et al, 2000; Martin-Henao
et al, 2000). The cells are captured by an open field
magnetic system using rare earth magnets and then washed
three times by capture and release. The anti-CD34 magnetic
bead complex is subsequently removed from cells by the
excess addition of an octapeptide (PR34
+
stem cell releasing
agent), which binds with high affinity to the epitope
recognized by the 9C5 anti-CD34 monoclonal antibody.
Studies have been carried out to evaluate the perform-
ance of these devices, many of which have utilized earlier
versions of the software running the devices (Stainer et al,
1998; Schumm et al, 1999; Croop et al, 2000; Despres et al,
2000; Hildebrandt et al, 2000; Martin-Henao et al, 2000).
None, however, has made a direct performance comparison
using identical PBSC products. Consequently, conclusions
regarding the relative merits of these devices are difficult to
make in view of differences between both the study centres
and, more importantly, the apheresis products processed on
the machines. In this study, we have directly compared
these two devices by selecting CD34
+
cells on each device
simultaneously. This was done by pooling and splitting
equally two PBSC harvests collected on successive days from
10 separate patients and processing half on the CliniMACS
device and half on the Isolex 300i device. The differences
observed in the performance of the two devices are now
reported with respect to ease of use, product purity, CD34
+
yield and residual T- and B-cell content. Additionally, we
have investigated the in vitro functional capacity of the two
CD34 purified products and report the differences observed
in the yield of colony-forming units (CFUs) and the
expansion of lineage-committed cells in liquid culture.
PATIENTS AND METHODS
Patients studied and PBSC collection. Following approval
from the local ethics committee and with informed patient
consent, 10 subjects were studied. Eight patients had a
diagnosis of multiple myeloma, of which five were mobilized
with cyclophosphamide (1Æ5 g/m
2
n¼3, 4 g/m
2
n¼2)
and granulocyte colony-stimulating factor (G-CSF), and
three were mobilized with ESHAP (etoposide, cisplatin,
cytarabine and methylprednisolone) and G-CSF as described
(Velasquez et al, 1994; Watts et al, 2000). One patient had
non-Hodgkin’s lymphoma and was mobilized with ESHAP
and G-CSF. The other subject was a normal donor for a
patient requiring an allograft and was mobilized with G-CSF
alone. Aphereses were performed on two successive days
using either the COBE Spectra (COBE Laboratories, Glou-
cester, UK) (n¼2) or the Baxter CS3000 (Baxter Health-
care, Deerfield, IL) (n¼8). Patients were eligible for the
study only if the combined CD34 count in the two
successive harvests was greater than 3 ·10
6
/kg.
Harvest split and cell processing. The first harvest was
diluted in an equal volume of autologous plasma and stored
overnight at room temperature in gas-permeable bags in
accordance with CliniMACS recommendations for harvest
storage, with the cell count not exceeding 2 ·10
8
/ml. The
next day, it was pooled with the second harvest and the
resultant combined harvest was mixed and then split in to
two equal volumes for CD34 processing. Both the Clini-
MACS and Isolex 300i machines were operated according to
their standard operating protocols and no allowance was
made for high CD34 target counts. The CliniMACS machine
was used in conjunction with the COBE 2991 cell washer.
Standard kits and reagents were used according to manu-
facturers’ guidelines. Version 2Æ5 software was used with
the Isolex 300i and version 2Æ1 software with the Clini-
MACS.
Flow cytometry. Flow cytometry was performed using an
EPICS Elite flow cytometer (Beckman-Coulter, High Wyco-
mbe, UK). Harvest and purified product CD34 cell numbers
were determined as described previously (Pollard et al,
1999). Phycoerythrin (PE)-conjugated anti-CD34 and flu-
orescein isothiocyanate (FITC)-conjugated anti-CD45 were
from Becton-Dickenson (San Jose, CA, USA). Harvest and
purified product T- and B-cell contents were determined
using a four-colour flow cytometric method. Briefly, 5 llof
apheresis harvest, diluted 1:20 in AB serum (approximately
1·10
6
cells), or 200 ll of product (also approximately
1·10
6
cells), was incubated for 30 min with PC5-conju-
gated anti-CD3, RD1 conjugated anti-CD4, FITC conjugated
anti-CD8 and energy-coupled dye (ECD) conjugated anti-
CD19 (Beckman-Coulter). Similar numbers of cells were
incubated with appropriate control antibodies. Samples
were then treated with Q-prep (Beckman-Coulter) to lyse red
cells and washed in phosphate-buffered saline (PBS) before
flow cytometry where a minimum of 50 000 events were
recorded. To determine retention of mouse immunoglobulin
on the surface of processed cells, 100 ll of product was
incubated for 15 min with a FITC-conjugated anti-mouse
immunoglobulin (Ig) (Dako, Ely, UK). Apoptotic cells in the
freshly purified products were detected by determining
Annexin V binding using a commercial kit (Annexin-
V-FLUOS Staining kit, Boehringer Mannheim, Germany)
in accordance with manufacturer’s instructions.
Colony assays. Colony-forming unit content of the har-
vest and product was assessed in semi-solid, methylcellu-
lose-based media (MethoCult H4230; Stem Cell
Technologies, Vancouver, Canada) with 20% added Iscove’s
modified Dulbecco’s medium (IMDM) (Life Technologies,
Paisley, UK), stem cell factor (SCF, 10 ng/ml) (Sigma, Poole,
UK), interleukin 3 (IL-3, 30 ng/ml), granulocyte/monocyte
colony-stimulating factor (GM-CSF, 25 ng/ml) (both from
Sandoz, Berne, Switzerland), G-CSF (25 ng/ml) (Amgen,
Thousand Oaks, CA, USA) and erythropoietin (EPO, 3 U/ml)
(Roche, Basel, Switzerland). Briefly, cells from the apheresed
harvests were suspended in 2Æ5 ml of media at
25 000 cells/ml and were cultured split between four
0Æ5 ml wells in a 24-well plate at 37C and 4% CO
2
. Cells
from the CD34-purified products were suspended in media
at a concentration of 500 cells/ml and were plated out
similarly. Colonies were counted on d 14.
Liquid culture of progenitor cells. CD34
+
-selected cells were
placed in culture in one well of a six-well plate on the day of
processing at a density of 1–2Æ5·10
5
/ml in IMDM, 20%
fetal calf serum (FCS) (Life Technologies) and 1% penicillin
118 M. J. Watts et al
2002 Blackwell Science Ltd, British Journal of Haematology 118: 117–123
and streptomycin (Sigma) at 37C and 5% CO
2
. To generate
erythroid cells, this medium was supplemented with
EPO (2 U/ml), SCF (20 ng/ml) and IL-3 (1 ng/ml). To
generate myeloid cells medium was supplemented with SCF
(20 ng/ml), IL-3 (20 ng/ml) and G-CSF (100 ng/ml).
During culture, cells were maintained at a density less than
1·10
6
/ml by dilution with medium supplemented with
growth factors whenever necessary. Cell counts and
estimations of viability using Trypan blue were performed
on d 0, 2, 4, 6 and 9 of the culture. On d 9 of the culture,
cytospin preparations demonstrated that cells in the eryth-
roid culture were predominantly at early to intermediate
normoblast stage and that cells in the myeloid culture were
predominantly at the myelocyte stage of differentiation
(data not shown).
Transendothelial migration assays. Assays were performed
as described (Yong et al, 1998). Briefly, 3 ·10
5
CD34-
selected cells were placed in a 3-lm Transwell filter (Costar,
Corning, NY) containing a confluent endothelial cell
monolayer. They were allowed to migrate overnight, with
100 ng/ml stromal-derived growth factor (SDF-1, Pepro-
Tech, London, UK) as a chemoattractant. At the end of the
experiment, transmigrated cells were recovered from the
lower compartment and set up in colony assays, as detailed
above. Percentage migration of granulocyte/macrophage-
CFU (CFU-GM) and erythroid burst-forming units (BFU-E)
was calculated from the colony content of aliquots of the cell
suspension originally seeded on to the filters.
RESULTS
Product CD34
+
cell purity and yield
Ten pairs of harvests were available for processing. Each
pair was pooled, gently mixed and then split equally. The
median total nucleated cell count in the resultant 20
harvests was 28 ·10
9
(range 15–56 ·10
9
) and the
median CD34
+
cell content was 1Æ8% (range 0Æ4–6Æ4%),
with no significant difference between matched pairs.
Following processing of one of each matched pair on each
of the two devices, the product obtained from the CliniMACS
machine was found to have significantly higher CD34
+
purity, although the yield of CD34
+
cells from the harvests
was similar for both machines (Table I). It was noted that in
the two harvest pairs in which the absolute CD34
+
cell
count was above 1Æ0·10
9
per individual split harvest, the
yield of CD34
+
cells was low from both machines (27% and
45% for the CliniMACS, and 40% and 42%, respectively, for
the Isolex 300i). The manufacturer’s guidelines for maxi-
mum capture of target CD34
+
cells on the CliniMACS device
(6 ·10
8
cells) was exceeded in these cases. In one other
case, CD34
+
cell yield was very low at 25%. On this
occassion, the CliniMACS column became blocked by DNA
gel half way through the procedure. In all other cases, the
yield of CD34
+
cells was above 40%.
Product T- and B-cell content
The products were analysed for T- and B-cell content. As
shown in Table II, the median T-cell content of the
CliniMACS product was significantly lower than that of
the Isolex 300i product. The difference in CD4 cell content
was also statistically significant, although the difference in
CD8 content only bordered on statistical significance. This
reduction in overall T-cell content represented a highly
significant median 0Æ9 log further reduction achieved with
the CliniMACS machine by comparison with the Isolex
300i. There was no statistically significant difference in the
B-cell content of the two products.
CliniMACS cells retain surface mouse Ig
As shown in Table III, and consistent with the mechanism
of cell processing, the iron oxide/dextran conjugated mouse
anti-CD34 antibody used to select the cells on the Clini-
MACS device was retained on the surface of the CD34
+
cells,
whereas this was not the case with cells purified on the
Isolex 300i device.
Viability and clonal expansion
The recovery of CFUs by each device was assessed by
comparing the number of CFUs obtained from the processed
product with the number of CFUs contained within the
unprocessed harvest. As shown in Table IV, median overall
CFU recovery was significantly higher in the Isolex 300i
product compared with the CliniMACS product.
Lineage-specific expansion of cells from the selected
products was additionally assessed in liquid culture medium
in eight of the paired products. Equal numbers of viable cells
from each product, as determined by Trypan blue exclusion,
were placed into liquid culture medium with growth factors
that support either myeloid or erythroid cell expansion. As
shown in Fig 1, in conditions that supported the expansion
of either myeloid or erythroid cells, the Isolex 300i product
generated significantly more cells at all time-points up to
d 9 than cells from the CliniMACS product. On d 9 of the
myeloid culture, mean expansion of cells from the Isolex
300i device was 55-fold (range 9–110) compared with a
35-fold (range 3–80) expansion of cells from the CliniMACS
device (P¼0Æ03). On d 9 of the erythroid expansion, Isolex
300I-processed cells had undergone a mean 155-fold (range
7–324) expansion whereas CliniMACS cells had expanded
73-fold (range 6–203) (P¼0Æ03). This difference is likely to
be explained by the apoptosis of a greater proportion of the
CliniMACS processed cells in the 48 h following the start of
the culture, as compared with the Isolex 300i processed
Table I. Purified product CD34 purity and yield (median and
range).
Machine CD34 purity (%) CD34 yield (%)
Isolex 300i 78 (60–93) 60 (40–84)
CliniMACS 90 (79–99) 64 (24–76)
P-value (t-test) 0Æ004 0Æ85
CD34
+
cell content of the harvest and product was
determined by flow cytometry. CD34
+
cell yield was deter-
mined by dividing the number of CD34
+
cells in the product
by the number of CD34
+
cells in the split harvest.
CliniMACS
(v.2Æ1) vs Isolex
300i (v2Æ5) 119
2002 Blackwell Science Ltd, British Journal of Haematology 118: 117–123
cells. Using Trypan blue exclusion as the determinant of cell
viability, on d 2 of the erythroid culture, compared with
d 0, the percentage viability of the CliniMACS-processed
cells had fallen by a median of 23Æ5% (range 11–60%). This
was a significantly greater drop than the median 13%
(range )8 to 57%) drop in viability observed with the Isolex
300I-processed cells (P¼0Æ04).
Further evidence that the CliniMACS products contained
an increased proportion of terminally damaged cells was
seen when the proportion of apoptotic cells in both products
was assessed using an annexin V binding assay. Cells that
exclude Trypan blue may nevertheless be committed to
apoptosis, and a proportion of these may be identified by
annexin V binding. As shown in Table V, the median
percentage of apoptotic cells in the CliniMACS product was
significantly higher than the Isolex 300i product.
The logarithmic expansion curves for both erythroid and
myeloid cultures were parallel following d 2 (Fig 1), sug-
gesting no difference in the proliferative potential of
surviving progenitor cells. Furthermore, there was no
significant difference found between the products from
either machine when the time and intensity of upregulation
Table II. Purified product T- and B-cell content (median and range).
Machine CD3
+
(%) CD4
+
(%) CD8
+
(%) Log CD3
+
depletion CD19
+
(%)
Isolex 300i 0Æ44 (0Æ06–0Æ76) 0Æ11 (0Æ01–0Æ34) 0Æ10 (0Æ00–0Æ44) 3Æ6(3Æ3–4Æ5) 0Æ03 (0Æ00–0Æ75)
CliniMACS 0Æ06 (0Æ01–0Æ39) 0Æ04 (0Æ00–0Æ18) 0Æ01 (0Æ00–0Æ18) 4Æ5(3Æ8–5Æ6) 0Æ12 (0Æ00–0Æ77)
P-value (t-test) 0Æ003 0Æ02 0Æ06 < 0Æ001 0Æ55
T- and B-cell contents of the product were determined using a four-colour flow cytometric assay. The log CD3
+
cell depletion was determined
by comparing the number of CD3
+
cells in the product with the number of CD3
+
cells in the harvest.
Table III. Purified product cell surface anti-CD34
immunoglobulin retention (median and range).
Machine
Cells retaining bound
mouse immunoglobulin (%)
Isolex 300i 0Æ5 (0–1Æ6)
CliniMACS 96 (67–99)
P-value (t-test) < 0Æ0001
Retention of anti-CD34 immunoglobulin was
determined with a flow cytometric assay utilizing
an anti-mouse Ig FITC-conjugated antibody.
Table IV. Purified product CFU yield (median and range).
Machine
Total CFU
yield (%)
BFU-E
yield (%)
CFU-GM
yield (%)
Isolex 300i 48 (18–77) 39 (10–63) 60 (24–95)
CliniMACS 38 (5–63) 26 (3–43) 53 (7–84)
P-value (t-test) 0Æ035 0Æ002 0Æ27
Colony-forming unit content (both BFU-E and CFU-GM) of har-
vest and product were determined by culture of aliquots of cells in
semi-solid methylcellulose culture. The CFU, BFU-E and CFU-GM
yields were determined by dividing the total number of CFUs in the
product by the total number of CFUs in the split harvest.
Fig 1. Myeloid and erythroid expansion in liquid culture.Equal
numbers of viable CD34
+
-selected progenitors, as assessed by Try-
pan blue dye exclusion, were placed in liquid culture medium that
supported either myeloid or erythroid lineage expansion. Viable
cells were counted on the indicated days and fold expansion over
input numbers was determined for CliniMACS and Isolex 300i
processed cells (mean ± SEM).
120 M. J. Watts et al
2002 Blackwell Science Ltd, British Journal of Haematology 118: 117–123
of the erythroid specific marker Glycophorin A during
erythroid expansion or the myeloid marker CD11b during
myeloid expansion was compared (data not shown).
Transendothelial migration assays
In three cases, the transendothelial migratory capacity of
cells from the CliniMACS and Isolex 300i products was
assessed. There was no significant difference in the trans-
migratory capacity of either CFU-GM or BFU-E derived from
either product. Percentage migration of CFU-GM from the
CliniMACS product was 7Æ7±0Æ5% compared with
10Æ8±3Æ7% for the Isolex 300i (mean ± SEM). Similar
results were obtained for BFU-E migration. This further
suggested that the functional performance of surviving
progenitor cells was equivalent.
DISCUSSION
This study is the first to compare directly the CliniMACS and
Isolex 300i clinical scale CD34
+
selection devices. By pooling
and splitting two PBSC harvests obtained on sequential days,
it was ensured that each device processed identical material.
We found that both devices generally worked reliably and
were straightforward to use. Although the time taken to
process the cells using the CliniMACS machine in conjunc-
tion with the COBE 2991 cell washer was about 1 h faster
than the Isolex 300i (approximately 160 min vs 220 min),
the latter machine had the advantage of being a single
platform device. Additionally, during one procedure the
CliniMACS column became blocked by DNA gel, something
that did not occur with the Isolex 300i machine, probably
due to the use of DNAase.
We found the CD34
+
purity of the CliniMACS product to
be significantly higher than that of the Isolex 300i product.
This finding concurs with other published studies that
evaluated the devices individually. These have reported
higher median CD34
+
purities in products from the Clini-
MACS device (97% (n¼71) in Schumm et al (1999) and
98% (n¼30) in Despres et al (2000) than from the Isolex
300i device (82% (n¼13) in Stainer et al (1998); 89%
(n¼51) in Martin-Henao et al (2000); 84% (n¼43) in
Hildebrandt et al (2000); and 85% (n¼30) in Croop et al
(2000). The reported median yields of CD34
+
cells have also
been higher for the CliniMACS device (71%, Schumm et al,
1999; 70%, Despres et al, 2000) than for the Isolex 300i
device (50%, Stainer et al, 1998; 58%, Martin-Henao et al,
2000; 51%, Hildebrandt et al, 2000; and 43%, Croop et al,
2000). However, when we compared the devices directly,
the yields were found to be similar for both machines at 60–
64%. This emphasizes the difficulty in directly comparing
machine evaluation studies performed by different centres
on different patient products with different devices utilizing
different software. It was of note that at CD34
+
input levels
between 1Æ0 and 1Æ5·10
9
, both machines suffered a
similar drop in yield, demonstrating similar saturation
points for CD34
+
recovery.
The median product CD34
+
purities obtained in this
study were at the low end of the ranges previously described
for both devices. This may be due to the use of pooled
harvests in this study, with half being stored overnight at
room temperature, as discussed later. Our experience of
processing fresh harvests, which have not been stored
overnight, is similar to that of others. In 55 consecutive
CD34 selection procedures performed during 2000 and
2001, median CD34
+
purity and yield were 95% and 67%,
respectively, for the CliniMACS device (n¼45), and 90%
and 57%, respectively, for the Isolex 300i device (n¼10).
Previous studies also report greater T-cell depletion with
the CliniMACS device (Schumm et al, 1999) than the Isolex
300i device (Stainer et al, 1998). Our data, which also
shows better T-cell depletion with the CliniMACS device,
confirm this finding. Although the CliniMACS device gave
more efficient T-cell depletion, there was no significant
difference in B-cell depletion between the two devices. It is
not clear why this should be, although it is possible that
inherent differences between T- and B-cell surface proteins
and carbohydrates cause different non-specific interactions
with the two columns, thus altering non-specific retention
of the two cell types.
When the functional capacity of the product was
investigated using a liquid culture ex vivo expansion
method, the CliniMACS product was found to generate
approximately half the number of erythroid and two-thirds
of the number of myeloid precursors at all time-points up to
d 9 compared with the Isolex 300i product. Additionally,
recovery of CFUs from the CliniMACS product was 21% less
than from the Isolex 300i product. This is despite more
CD34
+
cells being put in to both the liquid and semi-solid
cultures from the CliniMACS product than the Isolex 300i
product, because the CD34
+
purity of the CliniMACS
product was significantly higher. If correction had been
made for CD34
+
purity when cultures were set up, the
observed difference would have been even greater.
This finding suggests that in the CliniMACS product there
was a greater proportion of myeloid- and erythroid lineage-
specific CD34
+
cells that were either dead or viable with
respect to Trypan blue exclusion but nevertheless committed
to apoptosis. The reason for this is not clear, although several
possibilities arise. First, the CliniMACS device may non-
specifically retain dead cells or cells committed to apoptosis.
It has been reported that dead cells contaminating a harvest
may non-specifically bind the iron oxide/dextran conjugated
Table V. Purified product apoptotic cell content
(immediately post processing) (median and range).
Machine Annexin V-positive cells (%)
Isolex 300i 18 (12–34)
CliniMACS 28 (15–51)
P-value (t-test) 0Æ007
Aliquots of cells from the purified products were
incubated with annexin V-FLUOS in the appro-
priate binding buffer. Apoptotic cells binding
annexin V were subsequently identified using flow
cytometry.
CliniMACS
(v.2Æ1) vs Isolex
300i (v2Æ5) 121
2002 Blackwell Science Ltd, British Journal of Haematology 118: 117–123
antibody used with the CliniMACS machine (Schumm et al,
1999). Although the same non-specific interaction may
occur with the Isolex 300i device, viable CD34
+
cells are
released from the column with PR34
+
stem cell releasing
agent, which may release fewer non-specifically bound
apoptotic cells. Additionally, overnight storage of cells at
room temperature may have contributed to increasing the
proportion of apoptotic cells in the processed harvests,
consequently exacerbating this tendency. Although this is
the recommended method of storage of products to be run
on the CliniMACS device, when the nucleated cell count in a
fresh harvest is greater than 1Æ5·10
8
/ml, as was the case
in eight out of 10 of the overnight-stored harvests in this
study, it has been reported that overnight storage at room
temperature increases the proportion of apoptotic cells,
probably due to reduced product pH (Greer et al, 1997;
Ahmed et al, 2000).
An alternative explanation for the reduced functional
capacity of the CliniMACS product is that the mechanical
stresses involved during the CliniMACS processing may
irreversibly damage a proportion of the cells, which are
unaffected by the alternative mechanisms used by the Isolex
300i device. This explanation has been suggested by others
to explain similar findings of reduced viability in the
CliniMACS product (Fritsch et al, 2000), although possible
causes remain speculative.
Another possibility is that the different monoclonal
antibodies used select different subpopulations of CD34
+
cells, with the 9C5 antibody (used with the Isolex 300i
device) selecting a population with greater clonogenic
capacity than that selected by the QBEnd10 antibody (used
with the CliniMACS device). The existence of differences in
the expression of Class 2 Pasteurella glycoprotease-sensitive
CD34 epitopes and Class 3 cleavage-resistant CD34 epitopes
on subpopulations of haemopoietic progenitor cells has been
suggested before (Steen et al, 1996). The QBEnd10 and 9C5
antibodies, however, both recognize Class 2 CD34 epitopes
(Steen et al, 1996; Martin-Henao et al, 2000) and, although
we cannot rule it out, it seems unlikely that our results are
explained by differential subpopulation selection.
In most situations, the differences demonstrated in this
study in the functional capacity of the products from the
two machines will not matter. Surviving progenitor cells
have equivalent in vitro proliferative and migratory capacity
regardless of the machine used. However, where the
number of CD34
+
cells following selection is predicted to
be marginal (i.e. near to the previously determined thresh-
old for uncomplicated autograft engraftment of 2 ·10
6
/kg
(Watts et al, 1997; Watts et al, 2000) the choice of machine
used to process the harvest could become important.
Additionally, because this study demonstrates that a
proportion of processed CD34
+
cells that are intact and
resistant to Trypan blue are already committed to apoptotic
cell death, it is clear that determination of the number of
CD34
+
cells in the product alone is an insufficient assess-
ment of the functional capacity of the product. Additional
functional assays, such as CFU determination, performed in
parallel with immunophenotyping are essential to assess
fully the product’s suitability for transplantation.
ACKNOWLEDGMENTS
This study was funded in part by a research grant from
Baxter. Additionally, T.C.P.S. is supported by a personal
fellowship from the Medical Research Council (UK), and
A.K., K.Y. and D.C.L. are supported by a co-operative group
grant from the Medical Research Council (UK).
REFERENCES
Ahmed, F., Watts, M.J., Ings, S.J. & Flynn, M. (2000) Poor viability
of freshly apheresed PBSC harvests: implications for overnight
storage before cryopreservation or additional manipulative pro-
cedures. Blood,98, 380a (abstract 1639).
Ardeshna, K.M., Corney, C.P., Ings, S.J., Watts, M.J., Linch, D.C. &
Devereux, S. (2000) A clinically applicable method for the ex vivo
generation of antigen-presenting cells from CD34
+
progenitors.
Vox Sanguis,79, 46–52.
Cagnoni, P.J. & Shpall, E.J. (1996) Mobilization and selection
of CD34-positive hematopoietic progenitors. Blood Reviews,10,
1–7.
Croop, J.M., Cooper, R., Seshadri, R., Fernandez, C., Graves, V.,
Kreissman, S., Smith, F.O., Cornetta, K., Williams, D.A. &
Abonour, R. (2000) Large-scale mobilization and isolation of
CD34
+
cells from normal donors. Bone Marrow Transplanation,
26, 1271–1279.
Despres, D., Flohr, T., Uppenkamp, M., Baldus, M., Hoffmann, M.,
Huber, C. & Derigs, H.G. (2000) CD34
+
cell enrichment for
autologous peripheral blood stem cell transplantation by use of
the CliniMACs device. Journal of Hematotherapy and Stem Cell
Research,9, 557–564.
Devereux, S., Corney, C., Macdonald, C., Watts, M., Sullivan, A.,
Goldstone, A.H., Ward, M., Bank, A. & Linch, D.C. (1998) Fea-
sibility of multidrug resistance (MDR-1) gene transfer in patients
undergoing high-dose therapy and peripheral blood stem cell
transplantation for lymphoma. Gene Therapy,5, 403–408.
Fritsch, G., Scharner, D., Froschl, G., Buchinger, P., Peters, C.,
Matthes, S., Witt, V. & Gadner, H. (2000) Selection of CD34-
positive blood cells for allogeneic transplantation: approaches to
optimize D34-Cell recovery, purity, viability, and T-cell depletion.
Onkologie,23, 449–456.
Gratwohl, A., Passweg, J., Baldomero, H. & Urbano-Ispizua, A.
(2001) Hematopoietic stem cell transplantation activity in
Europe 1999. Bone Marrow Transplantation,27, 899–916.
Greer, M., Hami, L., Zumerchik, P., Green, T., Goodrich, R. & Shpall,
E.J. (1997) Viability and recovery of PBSC products stored at
room temperature is dependent on storage conditions. Blood,90,
290a (abstract 920).
Hildebrandt, M., Serke, S., Meyer, O., Ebell, W. & Salama, A. (2000)
Immunomagnetic selection of CD34
+
cells: factors influencing
component purity and yield. Transfusion,40, 507–512.
Kawano, Y., Takaue, Y., Watanabe, A., Takeda, O., Arai, K., Itoh,
E., Ohno, Y., Teshima, T., Harada, M., Watanabe, T., Okamoto,
Y., Abe, T., Kajiume, T., Matsushita, T., Ikeda, K., Endo, M.,
Kuroda, Y., Asano, S., Tanosaki, R., Yamaguchi, K., Law, P. &
McMannis, J.D. (1998) Partially mismatched pediatric trans-
plants with allogeneic CD34(
+
) blood cells from a related donor.
Blood,92, 3123–3130.
Martin-Henao, G.A., Picon, M., Amill, B., Querol, S., Gonzalez, J.R.,
Martinez, C., Martino, R., Ferra, C., Brunet, S., Granena, A.,
Sierra, J. & Garcia, J. (2000) Isolation of CD34
+
progenitor cells
from peripheral blood by use of an automated immunomagnetic
selection system: factors affecting the results. Transfusion,40,
35–43.
122 M. J. Watts et al
2002 Blackwell Science Ltd, British Journal of Haematology 118: 117–123
Pollard, Y., Watts, M.J., Grant, D., Chavda, N., Linch, D.C. &
Machin, S.J. (1999) Use of the haemopoietic progenitor cell count
of the Sysmex SE-9500 to refine apheresis timing of peripheral
blood stem cells. British Journal of Haematology,106, 538–544.
Reiffers, J., Cailliot, C., Dazey, B., Attal, M., Caraux, J. & Boiron, J.M.
(1999) Abrogation of post-myeloablative chemotherapy neutro-
penia by ex vivo expanded autologous CD34-positive cells. Lancet,
354, 1092–1093.
Schmitz, N., Linch, D.C., Dreger, P., Goldstone, A.H., Boogaerts,
M.A., Ferrant, A., Demuynck, H.M., Link, H., Zander, A. & Barge,
A. (1996) Randomised trial of filgrastim-mobilised peripheral
blood progenitor cell transplantation versus autologous bone-
marrow transplantation in lymphoma patients. Lancet,347,
353–357.
Schumm, M., Lang, P., Taylor, G., Kuci, S., Klingebiel, T., Buhring,
H.J., Geiselhart, A., Niethammer, D. & Handgretinger, R. (1999)
Isolation of highly purified autologous and allogeneic peripheral
CD34
+
cells using the CliniMACS device. Journal of Hematothera-
py,8, 209–218.
Stainer, C.J., Miflin, G., Anderson, S., Davy, B., McQuaker, I.G. &
Russell, N.H. (1998) A comparison of two different systems for
CD34
+
selection of autologous or allogeneic PBSC collections.
Journal of Hematotherapy,7, 375–383.
Steen, R., Tjonnfjord, G.E., Gaudernack, G., Brinch, L. & Egeland, T.
(1996) Differences in the distribution of CD34 epitopes on normal
and haemopoietic progenitor cells and leukaemic blast cells.
British Journal of Haematology,94, 597–605.
Urbano-Ispizua, A., Rozman, C., Martinez, C., Marin, P., Briones, J.,
Rovira, M., Feliz, P., Viguria, M.C., Merino, A., Sierra, J., Mazzara,
R., Carreras, E. & Montserrat, E. (1997) Rapid engraftment
without significant graft-versus-host disease after allogeneic
transplantation of CD34
+
selected cells from peripheral blood.
Blood,89, 3967–3973.
Velasquez, W.S., McLaughlin, P., Tucker, S., Hagemeister, F.B.,
Swan, F., Rodriguez, M.A., Romaguera, J., Rubenstein, E. &
Cabanillas, F. (1994) ESHAP: an effective chemotherapy regimen
in refractory and relapsing lymphoma: a 4-year follow-up study.
Journal of Clinical Oncology,12, 1169–1176.
Watts, M.J., Sullivan, A.M., Ings, S.J., Leverett, D., Peniket, A.J.,
Perry, A.R., Williams, C.D., Devereux, S., Goldstone, A.H. &
Linch, D.C. (1997) Evaluation of clinical scale CD34
+
cell puri-
fication: experience of 71 immunoaffinity column procedures.
Bone Marrow Transplantation,20, 157–162.
Watts, M.J., Ings, S.J., Leverett, D., MacMillan, A., Devereux, S.,
Goldstone, A.H. & Linch, D.C. (2000) ESHAP and G-CSF is a
superior blood stem cell mobilizing regimen compared to cyclo-
phosphamide 1.5 g/m
2
and G-CSF for pre-treated lymphoma
patients: a matched pairs analysis of 78 patients. British Journal of
Cancer,82, 278–282.
Yong, K.L., Watts, M., Shaun, T.N., Sullivan, A., Ings, S. & Linch,
D.C. (1998) Transmigration of CD34
+
cells across specialized and
non-specialized endothelium requires prior activation by growth
factors and is mediated by PECAM-1 (CD31). Blood,91, 1196–
1205.
CliniMACS
(v.2Æ1) vs Isolex
300i (v2Æ5) 123
2002 Blackwell Science Ltd, British Journal of Haematology 118: 117–123
... Granulocyte-monocyte colony-forming unit-(CFU-GM) assays were used to assess potency. These assays were set up by dilution of fresh HPC samples with Iscove's media and 2% human albumin solution (HAS), in the case of thawed samples adding warmed media dropwise, to prepare cell plating concentrations of 2Á5 9 10 4 /ml in methylcellulose media enriched with growth factors (StemMACS HSC-CFU complete with EPO, Miltenyi Biotec, Bisley, UK) (Watts et al, 2002(Watts et al, , 2008. Each sample was then dispensed in four 0Á5-ml aliquots in Costar 24-well plates (Sigma-Aldrich, Gillingham, UK) and colonies counted after a 14-d incubation at 37°C in a 5% C0 2 atmosphere, taking the mean of the four well readings as the colony number/well. ...
... Excess fresh PBSCs are not frequently available. To ensure that experiments were completed in a reasonable timescale, two products used were CD34-poor flow-through 'waste' fractions of two clinical CD34-purification procedures (Watts et al, 2002) run at UCLH (known to have low progenitor numbers), and the remaining cases were collections where there was an excess of progenitors above clinical requirement so that the surplus cells (which would otherwise have been discarded) could be used for these assays. In each >0Á5 9 10 9 /l (absolute neutrophil count [ANC]>0Á5) or platelet recovery >20 9 10 9 /l, at least 7 d after last transfusion (Plt >20 9 10 9 /l). ...
Article
Standard quality assurance (QA) of cryopreserved peripheral blood stem cells (PBSC) uses post-thaw viable CD34(+) cell counts. In 2013, concerns arose at Great Ormond Street Hospital (GOSH) about 8 patients with delayed engraftment following myeloablative chemotherapy with cryopreserved cell rescue, despite adequate post-thaw viable cell counts in all cases. Root cause analysis was undertaken; investigations suggested the freeze process itself was a contributing factor to suboptimal engraftment. Experiments were undertaken in which a single PBSC product was divided into three and cryopreserved in parallel using a control-rate freezer (CRF) or passive freezing method (-80°C freezer) at GOSH, or the same passive freezing at another laboratory. Viable CD34(+) counts were equivalent and adequate in each. Granulocyte-monocyte colony-forming unit assays demonstrated colonies from the products cryopreserved using passive freezing (both laboratories), but no colonies from products cryopreserved using the CRF. The CRF was shown to be operating within manufacturer's specifications with freeze-profile within acceptable limits. This experience has important implications for quality assurance for all transplant programmes, particularly those using cryopreserved products. The failure of post-thaw viable CD34(+) counts, the most widely used routine QA test available, to ensure PBSC function is of great concern and should prompt reassessment of protocols and QA procedures.
... Although in vitro cell manipulation can show promising results [32,38], it is more efficient and safe to avoid laboratory processing and differentiation of cells. MACS sorting of CD34+ cells is already utilised in the surgical treatment of haematological malignancies, so the technology is already translatable [39]. The within-surgery sorting of CD271+ AD-MSCs has great clinical potential as an autologous approach to improve the survival of adipose tissue, especially in large volume fat grafting during breast reconstruction. ...
Article
Full-text available
Background Autologous fat grafting is often a crucial aspect of reconstructive and aesthetic surgeries, yet poor graft retention is a major issue with this technique. Enriching fat grafts with adipose tissue-derived mesenchymal stem cells (AD-MSCs) improves graft survival—however, AD-MSCs represent a heterogeneous population. Selection of subpopulations of AD-MSCs would allow the targeting of specific AD-MSCs that may benefit fat graft survival more than the general AD-MSC population. Methods Human AD-MSCs were selected for the surface marker CD271 using magnetic-activated cell sorting and compared to the CD271 negative phenotype. These subpopulations were analysed for gene expression using Real-Time qPCR and RNA sequencing; surface marker characteristics using immunostaining; ability to form tubules when cultured with endothelial cells; and gene and protein expression of key angiogenic mediators when cultured with ex-vivo adipose tissue. Results Human AD-MSCs with the surface marker CD271 express angiogenic genes at higher levels, and inflammatory genes at lower levels, than the CD271− AD-MSC population. A greater proportion of CD271+ AD-MSCs also possess the typical complement of stem cell surface markers and are more likely to promote effective neoangiogenesis, compared to CD271− AD-MSCs. Conclusion Enriching grafts with the CD271+ AD-MSC subpopulation holds potential for the improvement of reconstructive and aesthetic surgeries involving adipose tissue.
... Given limited initial experience with positive CD34+ selection with use of the CliniMACS Plus, a conservative SE estimate of 0.30 was used based on the lower end of published ranges. 10 Such conservative estimates potentially overestimate the blood volumes to process, which consequently confers the advantage of avoiding undercollection of donors. ...
Article
Background: Cellular therapies using engineered T cells, haploidentical transplants, and autologous gene therapy are increasing. Specified CD3+ or high CD34+ doses are typically required for subsequent manufacturing, manipulation, or CD34+ selection. Simple, practical, and reliable lymphocyte and hematopoietic progenitor cell (HPC) collection algorithms accounting for subsequent CD34+ selection have not been published. Study design and methods: In this analysis of 15 haploidentical donors undergoing tandem lymphocyte and HPC collections, we validated one-step, practical prediction algorithms (Appendix S1, available as supporting information in the online version of this paper) that use conservative facility-specific collection efficiencies, CD34+ selection efficiency, and donor-specific peripheral counts to reliably achieve the target CD3+ and CD34+ product doses. These algorithms expand on our previously published work regarding predictive HPC collection algorithms. Results: Ninety-three percent of lymphocyte and 93% of CD34+ collections achieved the final target CD3+ and CD34+ product dose when our algorithm-calculated process volumes were used. Linear regression analysis of our algorithms for CD3+, preselection CD34+, and postselection CD34+ showed statistically significant models with R2 of 0.80 (root mean square error [RMSE], 31.3), 0.72 (RMSE, 385.7), and 0.56 (RMSE, 326.0), respectively, all with p values less than 0.001. Conclusion: Because achievement of CD3+ or CD34+ dose targets may be critical for safety and efficacy of cell therapies, these simple, practical, and reliable prediction algorithms for lymphocyte and HPC collections should be very useful for collection facilities.
... [16][17][18] The CliniMACS system is able to achieve a 5-log T cell depletion while retaining a median viability of 98%, and in direct comparison with ISOLEX 300i the CliniMACS system yielded a product with higher CD34+ purity (90% vs 78%, P = 0.004), and lower median T-cell content (0.06% vs 0.44%; P = 0.003). 19 The CliniMACS CD34 Reagent System was approved by the FDA for the use of GVHD prevention in patients with acute myeloid leukemia (AML) in the first remission receiving a transplant from an HLA-identical sibling donor in January 2014, and remains the only FDA approved to CD34+ selection system for use in HCT. The CliniMACS system can also be used to selectively deplete CD3+ T cells, CD19+ B cells, and T cell receptor αβ positive (TCRαβ + ) T cells. ...
Article
Graft‐versus‐host (GVHD) is an important cause of morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT). As donor T cells are recognized as key drivers of GVHD, some approaches to prevent GVHD have focused on T cell depletion of the allograft. In this review we summarize methods and outcomes of ex vivo T cell depleted (TCD) HCT with a focus on CD34+ selection. This platform is efficacious in preventing acute and chronic GVHD across a wide range of hematologic malignancies, and with the exception of chronic myeloid leukemia, is not associated with adverse relapse or survival outcomes compared to conventional GVHD prophylaxis platforms. In retrospective comparisons recipients of CD34+ selected HCT have higher rates of GVHD‐free relapse‐free survival (GRFS) than conventional HCT counterparts. Although CD34+ selected allografts require myeloablative and antithymocyte‐globulin based conditioning to support engraftment, abrogation of calcineurin inhibitors and methotrexate in this approach reduces its toxicity such that it can be considered in select older and more comorbid patients who could benefit from ablative HCT. A trial comparing GVHD prophylaxis regimens (BMT CTN 1301, NCT 02345850) has completed accrual and will be the first to compare CD34+ selected HCT with conventional HCT in a randomized prospective setting. Its findings have a potential to establish CD34+ selected HCT as a new standard‐of‐care platform for GVHD prevention.
... During the 6 day storage we collected data about percent of 7-AAD-cells, absolute number of viable CD34+ cells and % of early apoptotic cells. Watts et al. reported a median 28% (range: 15-51%) of apoptotic CD34+ cells in fresh CliniMACS products, explaining that higher apoptosis may be due to overnight storage of harvests at room temperature [28]. In contrast, we detected fewer apoptotic cells (4.9%) in the fresh CD34+ enriched cell products, which were isolated from leukapheresis products after overnight storage at 2-8 • C. Notably, further storage of CD34+ enriched cell products at 2-8 • C did not significantly change the percent of apoptotic CD34+ cells. ...
Article
Durable engraftment of transplanted CD34+ cells largely depends on the quality of the cell product. Limited data are currently available about extended storage of immunoselected CD34+ cells. The aim of our study was to assess the stability of CD34+ cell product with the cells stored in high concentration (80×10(6) in 6mL) in small bags intended for cell implantation. Cell products were prepared by leukapheresis and immunoselection (Clinimacs(plus) procedure) from 13 patients with chronic dilated cardiomyopathy. CD34+ cell products were stored at 2-8°C and analyzed at time 0 (fresh products), 24, 48h, 4 and 6 days. Product viability, absolute number of viable CD34+ cells and apoptosis were determined by flow cytometry. Microbiological contamination of the cell products was tested by BACTEC system. The mean viability of CD34+ cells decreased by 2.7% within 24h, by 13.4% within 48h and by 37.5% within 6 days. The mean recovery of viable CD34+ cells was 91.1%, 74.8%, 66.3% and 56.2% at 24, 48h, 4 and 6 days, respectively. The mean fraction of early apoptotic cells in fresh and stored products was 4.9±3.5% at 0h, 5.9±3.8% at 24h, 4.2±3.1% at 48h, 6.3±2.6% at 4 days and 9.3±4.6% at 6 days. All products were negative for microbial contamination.
... We further analyzed the cellular composition of Auto-CD34 + HPC lots based on the device used for CD34 + cell selection (Supplementary Appendix S1, Figure S4), including purity of Auto-CD34 + HPC, viability, recovery of CD34 + cells from the apheresis collection, and log TCD. Products manufactured on CliniMACS had higher viable CD34 + cell purity and were more highly depleted of CD3 + cells than those processed on Isolex (Supplementary Appendix S1, Figure S4), consistent with historic data [8][9][10][11]. The median recovery of CD34 + cells from apheresis collections and the median viability for Auto-CD34 + HPC were similar for the 2 devices. ...
Article
Full-text available
To ensure comparable grafts for autologous HCT in NIAID-sponsored IND protocols for multiple sclerosis (HALT-MS) and systemic sclerosis (SCOT), a DMF approach to control manufacture was implemented, including a common Master Production Batch Record, and site-specific SOPs with "Critical Elements". We assessed comparability of flow cytometry and controlled rate cryopreservation among sites, and stability of cryopreserved grafts, using hematopoietic progenitor cells (HPC) from healthy donors. Auto-CD34+HPC graft specifications included ≥70% viable CD34+ cells before cryopreservation. For the two protocols, 110 apheresis collections were performed; 121 lots of Auto-CD34+HPC were cryopreserved; 107 of these (88.4%) met release criteria. Grafts were infused at median 25 (range 17-68) days post-apheresis for HALT-MS (n=24), and 25 (14-78) days for SCOT (n=33). Subjects received pre-cryopreservation doses of median 5.1 (range 3.9-12.8) x 10(6) viable CD34+ cells/kg for HALT-MS, and 5.6 (2.6-10.2) x 10(6) viable CD34+ cells/kg for SCOT. Recovery of granulocytes occurred at median 11 (range 9-15) days post-HCT for HALT-MS, and 10 (8-12) days for SCOT, independent of CD34+ cell dose. Subjects received their last platelet transfusion at median 9 (range 6-16) days for HALT-MS, and 8 (6-23) days for SCOT; higher CD34+/kg doses were associated with faster platelet recovery. Stability testing of cryopreserved healthy donor CD34+ HPC over 6 months of vapor phase LN2 storage demonstrated consistent 69-73% recovery of viable CD34+ cells. Manufacturing of Auto-CD34+HPC for the HALT-MS and SCOT protocols was comparable across all sites and supportive for timely recovery of granulocytes and platelets.
... Briefly, iron-containing beads coated with anti-CD34 antibodies are mixed with the autologous graft product and the antibody-bound cells are isolated magnetically using semiautomated clinical scale devices. 14 This method has been tested on autologous graft products obtained from patients with breast cancer, neuroblastoma, myeloma, and lymphoma. [15][16][17][18] To date, there has only been one randomized controlled clinical trial evaluating the efficacy of an ex vivo purging technology. ...
Article
Full-text available
Jovian J Tsang,1,2 Harold L Atkins2,3 1Department of Biochemistry, University of Ottawa, 2Cancer Therapeutics, Ottawa Hospital Research Institute, 3Blood and Marrow Transplant Program, The Ottawa Hospital, Ottawa, ON, Canada Abstract: Hematological malignancies are treated with intensive high-dose chemotherapy, with or without radiation. This is followed by hematopoietic stem cell (HSC) transplantation (HSCT) to rescue or reconstitute hematopoiesis damaged by the anticancer therapy. Autologous HSC grafts may contain cancer cells and purging could further improve treatment outcomes. Similarly, allogeneic HSCT may be improved by selectively purging alloreactive effector cells from the graft rather than wholesale immune cell depletion. Viral agents that selectively replicate in specific cell populations are being studied in experimental models of cancer and immunological diseases and have potential applications in the context of HSC graft engineering. This review describes preclinical studies involving oncolytic virus strains of adenovirus, herpes simplex virus type 1, myxoma virus, and reovirus as ex vivo purging agents for HSC grafts, as well as in vitro and in vivo experimental studies using oncolytic coxsackievirus, measles virus, parvovirus, vaccinia virus, and vesicular stomatitis virus to eradicate hematopoietic malignancies. Alternative ex vivo oncolytic virus strategies are also outlined that aim to reduce the risk of relapse following autologous HSCT and mitigate morbidity and mortality due to graft-versus-host disease in allogeneic HSCT. Keywords: hematopoietic stem cells, oncolytic virus, hematopoietic stem cell transplantation, stem cell graft purging, hematopoietic malignancy, graft vs host disease
Chapter
This authoritative new book provides a comprehensive overview of diagnostic and therapeutic strategies in hematopoietic cell transplantation, explaining key concepts, successes, controversies and challenges. The authors and editors discuss current and future strategies for major challenges, such as graft-versus-host-disease, including new prophylaxis and treatments. They also discuss long-term complications, such as second malignancies and cardiovascular complications. Chapters are written by leading world experts, carefully edited to achieve a uniform and accessible writing style. Each chapter includes evidence-based explanations and state-of-the-art solutions, providing the reader with practice-changing advice. Full reference lists are also supplied to facilitate further exploration of each topic. Each copy of the printed book is packaged with a password, giving readers online access to all text and images. This inspiring resource demystifies both the basics and subtleties of hematopoietic cell transplantation, and is essential reading for both senior clinicians and trainees.
Article
Aim: The purpose of in vitro expansion of cord blood is to enhance the engrafting potential, and reconstruct the hematopoietic function, while engraftments have an important association with the large numbers of hematopoietic progenitor and homing potential. We aimed to explore the effect of interleukin (IL)-6/soluble interleukin 6 receptor (sIL-6R) on the expansion of the mononuclear cells derived from umbilical cord blood ex vivo and the expression of CXCR4 and CD49d after expansion of cord blood. Methods: Experiments were performed at Hematological Laboratory of Guangzhou First People' Hospital of Guangzhou Medical College from June 2005 to March 2006. 1 The 10 cord blood samples provided by Department of Gynaecology and Obstetrics of Guangzhou First People' Hospital was obtained from full-term healthy neonate. Informed consents were obtained from puerperants, and the experiment was ratified by Medical Ethics Commitee. 2 Human fresh cord blood mononuclear cells were cultured in serum-free and stroma-free medium containing different combination of cytokine for 7 days. According to the different combination of cytokine, the experiment was divided into four groups: control, SFT group (stem cell factor+FLT3+thrombopoietin, SCF+FL+TPO), SFT6 group (SCF+FL+TPO+IL-6), SFT6s group (SCF+FL+TPO+IL-6/sIL-6R). In line with the different concentrations of sIL-6R, the SFT6s group were further divided into four groups: 50 μ g/L group, 100 μ g/L group, 500 μ g/L group and 1000 μ g/L group. 3 The total cells were counted, CD34+ cells and CD34+CXCR4+, CD34+CD49d+ cells were assayed immediately and at day 7. Results: 1 Numbers of total nucleated cells and CD34+ cells were more in the SFT, SFT6, SFT6s group than in the control group (P < 0.05). There was no significant difference in the SFT, SFT6, SFT6s groups (P > 0.05). 2 Levels of CD49d, CXCR4 on expanded CD34+ cells from cord blood could be upregulated in the SFT, SFT6, SFT6s group compared to the control group, and there was no significant difference in the SFT, SFT6, SFT6s groups (P > 0.05). 3 The expansion and expression of CD49d, CXCR4 on expanded CD34+ cells from cord blood were higher in the 500, 1000 μ g/L SFT6s groups than in the SFT, SFT6s and 100 μ g/L SFT6s groups (P < 0.05), but there was no significant difference between 500 μ g/L SFT6s and 1000 μ g/L SFT6s groups (P > 0.05). Conclusion: The addition of IL-6/sIl-6R to SCF+FL+TPO combination can enhance expansion and expression of CD49d and CXCR4 of mononuclear cells derived from umbilical cord blood, but this effect depends on the concentration of sIL-6R.
Chapter
This chapter focuses on strategies for identification and separation of human hemopoietic stem cells (HSCs) and lineage-committed progenitors. It also discusses approaches to HSC expansion and recent clinical experience with ex vivo manipulated hemopoietic cells. During the last decade, there has been great interest in the development of methods for manipulating the growth and development of human HSCs and progenitor stem cells for therapy. These initiatives include the generation of committed progenitor cells and myeloid precursors for transplantation, expansion of HSCs from hemopoietic tissues, such as umbilical cord blood (CB) to increase safety and applicability of hemopoietic cell transplantation, and the use of HSCs as vehicles for gene therapy. Optimal methods for manipulating human HSCs and their progeny for any therapeutic application will arise from a comprehensive understanding of the molecular regulatory mechanisms that control stem cell fate and hemopoietic cell differentiation. Even though it is well recognized that the hemopoietic microenvironment plays a key role in these processes, the exact molecules responsible for these noncell autonomous aspects of HSC regulation remain to be elucidated. Accordingly, a major challenge for the field is to define the molecular "parts lists" for both stem and progenitor cell populations and the supporting microenvironment and moreover, to establish how these individual components interact to form regulatory pathways and networks.
Article
Full-text available
We have prospectively evaluated the feasibility and results of the biotin-avidin immunoadsorption method (Ceprate SC system) for a phase I/II study of T-cell depletion of granulocyte colony-stimulating factor (G-CSF) mobilized peripheral blood progenitor cells (PBPC) for allogeneic transplantation. Twenty consecutive patients, median age, 40 years (21 to 54) and diagnoses of chronic myeloid leukemia in chronic phase (n = 5), acute myeloblastic leukemia (n = 7), acute lymphoblastic leukemia (n = 2), chronic myelomonocytic leukemia (n = 1), refractory anemia with excess of blasts in transformation (n = 3), histiocytosis X (n = 1), and chronic lymphocytic leukemia (n = 1), were conditioned with cyclophosphamide (120 mg/kg) and total body irradiation (13 Gy; 4 fractions). HLA identical sibling donors received G-CSF at 10 microg/kg/d subcutaneously (SC); on days 5 and 6 (19 cases) and days 5 to 8 (1 case) donors underwent 10 L leukapheresis. PBPC were purified by positive selection of CD34+ cells using immunoadsorption biotin-avidin method (Ceprate SC) and were infused in the patients as the sole source of progenitor cells. No growth factors were administered posttransplant. The median recovery of CD34+ cells after the procedure was of 65%. The median number of CD34+ cells infused in the patients was 2.9 (range, 1.5 to 8.6) x 10(6)/kg. The median number of CD3+ cells administered was 0.42 x 10(6)/kg (range, 0.1 to 2). All patients engrafted. Neutrophil counts >500 and >1,000/microL were achieved at a median of 14 days (range, 10 to 18) and 15 days (range, 11 to 27), respectively. Likewise, platelet counts >20,000 and >50,000/microL were observed at a median of 10 days (range, 6 to 23) and 17 days (range, 12 to 130), respectively. Graft-versus-host disease (GVHD) prophylaxis consisted of cyclosporine plus methylprednisolone. No patient developed either grade II to IV acute or extensive chronic GVHD. After a median follow-up of 7.5 months (range, 2 to 22) three patients have relapsed, and one of them is again in hematologic and cytogenetic remission after infusion of the donor lymphocytes. Two patients died in remission: one on day +109 of pulmonary aspergillosis and the other on day +251 of metastasic relapse of a previous breast cancer. Sixteen of the 20 patients are alive in remission after a median follow-up of 7.5 months (range, 2 to 22). In conclusion, despite the small number of patients and limited follow-up, it appears that this method allows a high CD34+ cell recovery from G-CSF mobilized PBPC and is associated with rapid engraftment without significant GVHD, and with low transplant related mortality.
Article
Full-text available
Seventy-one mobilised PBSC collections were subject to CD34+ cell purification using the CEPRATE SC stem cell concentration system. The overall median purity of CD34+ cells was 69% (6-93%). CD34+ cell, and GM-CFC recoveries were 52% (8-107%) and 36% (3-118%). Purity was logarithmically related to the input percentage of CD34+ cells and starting requirements were established of 1% CD34 cell content for optimal purity and a minimum of 2 x 10(6)/kg CD34+ cells to ensure recovery of our minimum engraftment threshold of 1 x 10(6)/kg CD34+ cells. Reduction of the washing steps reduced non-specific cell losses and shortened the procedure but did not affect progenitor cell recovery. Purified CD34+ cells were reinfused following high-dose therapy in 35 patients. The median time to neutrophil recovery of 0.5 x 10(9)/l was 12 (10-23) days and to the attainment of platelet independence was 13 (7-100) days. The risks of delayed platelet recovery were related to the CD34+ cell dose infused and were identical to the risks when non-purified PBSC collections were used. In conclusion, purification of CD34+ cells using the CEPRATE device is reliable and the purified product results in prompt engraftment. The cell losses that occur do however restrict its use in many patients.
Article
Full-text available
We have performed a pilot study of MDR-1 gene transfer in patients receiving CD34-selected peripheral blood stem cell (PBSC) transplant for lymphoma. To ensure minimum engraftment thresholds and facilitate CD34 purification, mobilisation of > 2 x 10(6) CD34 cells/kg was a condition for recruitment. Of 11 patients counselled for study entry, only five achieved this target in a single apheresis. In three consenting patients, purified CD34 cells were exposed to A12M1 MDR-1 retroviral supernatant for 6 h, cryopreserved then thawed and readministered following ablative chemotherapy. No delay in engraftment was observed, although one patient received additional back-up cells. Gene transfer was demonstrated by polymerase chain reaction (PCR) for vector-derived MDR-1 cDNA sequence in all cases. Analysis of peripheral blood and bone marrow cells after transplant has, however, shown no evidence of in vivo gene transfer with a follow-up of 12, 15 and 18 months. The effect of MDR-1 substrate drugs has not yet been tested as all patients remain in clinical and radiological remission of their lymphoma. These results confirm the difficulty of achieving in vivo gene transfer in human haemopoietic cells and indicate major logistical constraints in PBSC mobilisation in patients with relapsed and resistant disease in whom initial studies are appropriate.
Article
Stepping motors are widely used for various electric instruments. It is necessary for the optimum design to analyze the dynamic characteristics accurately. Our method, which is the 3-D finite element method with edge elements taking into account the rotation of the armature, has been expanded to analyze the dynamic characteristics of a stepping motor for a wristwatch excited from square pulse voltage source. The validity of our method is confirmed by experiments
Article
This study attempted to determine the efficacy of the combination of etoposide (VP-16), methyl-prednisolone, and cytarabine (Ara-C) with or without cisplatin in relapsing and refractory adult lymphoma patients. The first 63 patients were randomized to receive VP-16 40 mg/m2/d for 4 days, methylprednisolone 500 mg intravenously daily for 5 days, and Ara-C 2 g/m2 intravenously over 2 to 3 hours on day 5 with or without cisplatin 25 mg/m2 IV administered by 24-hour infusion for 4 days (ESHA +/- P). Markedly different responses between ESHA (33%) and ESHAP (75%) led to deletion of the ESHA arm. A total of 122 patients on the ESHAP regimen were studied. Forty-five patients (37%) attained a complete remission (CR) and 33 (27%) attained a partial remission (PR), for a total response rate of 64%. The median duration of CR was 20 months, with 28% of remitters still in CR at 3 years. The overall median survival duration was 14 months; the survival rate at 3 years was 31%. Overall time to treatment failure (TTF) showed 10% of all patients to be alive and disease-free at 40 months. Response and survival rates were similar in patients with low-grade (n = 34), intermediate-grade (n = 67), transformed (n = 18), and high-grade (n = 3) lymphoma. The most significant factors for response and survival by multivariate analysis were the serum lactic dehydrogenase (LDH) level, tumor burden, and age (when analyzed as a continuous variable), while prior CR was highly significant by univariate analysis. A significant difference in survival was noted for patients with normal LDH levels and low- or intermediate-tumor burden or patients with low tumor burden and elevated LDH levels (55% 3-year survival rate) versus patients with elevated LDH levels and intermediate or high tumor burden (< 20%). Major toxicities included myelosuppression, with a median granulocyte count of 500/microL and platelet count of 70,000/microL. ESHAP was found to be an active, tolerable chemotherapy regimen for relapsing and refractory lymphoma. Applying a prognostic model based on tumor burden and serum LDH level shows significant differences in survival in this patient population.
Article
A randomised trial comparing filgrastim-mobilised peripheral blood progenitor cell (PBPC) transplants with autologous bone marrow transplantation (ABMT) for haematopoietic stem cell support has not been done. We compared the effects of filgrastim-mobilised PBPC or autologous bone marrow reinfused to lymphoma patients after high-dose chemotherapy in a prospective randomised multicentre trial. The trial was done at six centres in three European countries. After high-dose chemotherapy (carmustine, etoposide, cytarabine, and melphalan [BEAM protocol]) 58 patients with advanced Hodgkin's disease or high-grade non-Hodgkin lymphoma received either filgrastim-mobilised PBPC (n = 27) or bone marrow (n = 31) for haemopoietic reconstitution. The median number of days with platelet transfusions after grafting was 6 in the PBPC transplantation group and 10 in the ABMT group (estimate of treatment difference 5 days, 95% CI 3-7 days). Time to platelet recovery above 20 x 10(9)/L was 16 days in the PBPC transplantation group and 23 days in the ABMT group (p = 0.02). Time to neutrophil recovery above 0.5 x 10(9)/L was also reduced in the PBPC transplantation group (11 vs 14 days, p = 0.005). Patients randomised to PBPC transplantation needed fewer red blood cell transfusions (two vs three, p = 0.002) and spent less time in hospital (17 vs 23 days, p = 0.002). Early post-transplant morbidity and mortality as well as overall survival (median follow-up 311 days) were similar in both groups. There was no notable toxicity ascribed to filgrastim administration or the leucapheresis procedures. In patients with lymphoma treated with high-dose chemotherapy, reinfusing filgrastim-mobilised PBPC instead of autologous bone marrow significantly reduced the number of platelet transfusions, the time to platelet and neutrophil recovery, and led to earlier discharge from hospital.
Article
The CD34 molecule expressed on haemopoietic progenitor cells contains a large number of epitopes whose expression may be related to the maturation or function of the cells. Monoclonal antibodies specific for different epitopes have been reported to detect different numbers of CD34⁺ leukaemic blast cells. We wanted to confirm this observation and study whether parallel findings could be observed for normal haemopoietic progenitor cells. The cells were immunophenotyped by flow cytometry with a series of monoclonal antibodies reactive with different CD34 epitopes. Class III epitopes (resistant to enzymatic cleavage with neuraminidase, chymopapain and a glycoprotease from Pasteurella haemolytica) showed a broader distribution on normal haemopoietic progenitor cells and leukaemic blast cells than class I epitopes (sensitive to cleavage with all three enzymes) and class II epitopes (sensitive to degradation with glycoprotease and chymopapain, and resistant to neuraminidase). The subpopulation of normal progenitor cells which exclusively expressed class III epitopes had flow cytometric characteristics compatible with mature myeloid progenitor cells, whereas class I, II and III epitopes were equally expressed on cells enriched for immature subsets. No discordant epitope expression could be observed for the more immature leukaemias (AML-M0/1) and a higher percentage of the more mature leukaemic blast cells (AML-M3 and AML-M4/5) expressed class III epitopes compared to the percentage expressing class I and II epitopes.
Article
High-dose chemotherapy with autologous hematopoietic progenitor-cell support is increasingly used for the treatment of hematologic malignancies and solid tumors. Over the last few years, the major source of progenitor cells for clinical use has shifted from bone marrow to peripheral blood. The current approaches on peripheral blood progenitor-cell mobilization and collection is examined. The isolation of CD34-positive cells from peripheral blood progenitor-cell grafts for tumor purging is the autologous transplant setting and for T-cell depletion in the allogeneic transplant setting is also discussed.
Article
The transmigration of hematopoietic progenitor cells (HPCs) across vascular endothelium is a critical step in the homing of transplanted stem cells, but the molecular basis for this is unknown. We used mobilized peripheral blood CD34(+) selected cells and cultured bone marrow microvascular (BMECs) and human umbilical vein endothelial cells (HUVECs) to investigate the adhesion and transendothelial migration of HPCs. Colony-forming cells (CFCs) in freshly isolated CD34(+) cells showed high levels of adhesion to both forms of endothelium (28% +/- 4% and 38% +/- 6% of granulocyte-macrophage colony-forming cells [GM-CFCs] adhering to HUVECs and BMECs, respectively), but were unable to migrate to any significant extent across either (1.0% +/- 0.3% and 1.1% +/- 0.6% of GM-CFCs migrating across HUVECs and BMECs, respectively). Greater than 95% of peripheral blood CD34(+) cells are in G0/G1 of the cell cycle, but after 48 to 72 hours of stimulation with growth factors (interleukin-3 [IL-3] 12 ng/mL, stem cell factor 10 ng/mL, and IL-6 10 ng/mL), 28% +/- 5% of cells were in S+G2/M. Growth factor stimulation had no effect on the adhesion of mobilized CFCs but resulted in enhanced migration of these cells (9.8% +/- 1.6% and 12. 6% +/- 3.1% of GM-CFCs migrating across HUVECs and BMECs, respectively; P < .01, n = 6). Assessment of cell proliferation by the 3H-thymidine suicide method showed that, whereas 11.7% +/- 3.3% of proliferating CFCs transmigrated across endothelium, only 1.3% +/- 0.3% of nonproliferating CFCs did so (P < .05, n = 5). Transmigration of growth factor-activated CFCs was inhibited by anti-CD18 monoclonal antibody (MoAb; 50% +/- 18% inhibition) and by anti-platelet endothelial cell adhesion molecule-1 (PECAM-1) MoAb (70.8% +/- 7.1% inhibition; P < .05, n = 3). IL-1 stimulation of HUVECs had no significant effect on CD34(+) cell transmigration, but caused marked enhancement of neutrophil migration. Stem cell homing may depend, in part, on the ability of local cytokines to upregulate the transmigratory ability of these cells. The transmigration of HPCs shares at least some molecular pathways with that of mature cells (CD18 and PECAM-1), but is differently affected by endothelial activation.