ArticlePDF Available

Quantitative magnetic resonance and SPECT imaging for macrophage tissue migration and nanoformulated drug delivery

Authors:

Abstract

We posit that the same mononuclear phagocytes (MP) [bone marrow (BM) and blood monocytes, tissue macrophages, microglia, and dendritic cells] which serve as targets, reservoirs, and vehicles for HIV dissemination, can be used as vehicles for antiretroviral therapy (ART). Toward this end, BM macrophages (BMM) were used as carriers for nanoparticle-formulated indinavir (NP-IDV), and the cell distribution was monitored by single photon emission computed tomography (SPECT), transverse relation time (T2)* weighted magnetic resonance imaging (MRI), histology, and gamma-scintillation spectrometry. BMM labeled with super paramagnetic iron oxide and/or 111indium oxine were infused i.v. into naïve mice. During the first 7 h, greater than 86% of cell label was recorded within the lungs. On Days 1, 3, 5, and 7, less than 10% of BMM were in lungs, and 74-81% and 13-18% were in liver and spleen, respectively. On a tissue volume basis, as determined by SPECT and MRI, BMM densities in spleen and liver were significantly greater than other tissues. Migration into the lymph nodes on Days 1 and 7 accounted for 1.5-2% of the total BMM. Adoptive transfer of BMM loaded with NP-IDV produced drug levels in lymphoid and nonlymphoid tissues that exceeded reported therapeutic concentrations by 200- to 350-fold on Day 1 and remained in excess of 100- to 300-fold on Day 14. These data show real-time kinetics and destinations of macrophage trafficking and demonstrate the feasibility of monitoring macrophage-based, nanoformulated ART.
Quantitative magnetic resonance and SPECT imaging
for macrophage tissue migration and nanoformulated
drug delivery
Santhi Gorantla,*
,
Huanyu Dou,*
,
Michael Boska,*
,†,
Chris J. Destache,
§
Jay Nelson,*
,†,‡
Larisa Poluektova,*
,
Barett E. Rabinow,
Howard E. Gendelman,*
,†,1
and R. Lee Mosley*
,
*Center for Neurovirology and Neurodegenerative Disorders, Departments of
Pharmacology and Experimental
Neuroscience and
Radiology, University of Nebraska Medical Center, Omaha;
§
School of Pharmacy and Health
Professions, Creighton University, Omaha, Nebraska; and
Baxter Healthcare Corporation, Round Lake, Illinois
Abstract: We posit that the same mononuclear
phagocytes, bone marrow (BM) and blood mono-
cytes, tissue macrophages, microglia, and den-
dritic cells, which serve as targets, reservoirs, and
vehicles for HIV dissemination, can be used as
vehicles for antiretroviral therapy (ART). Toward
this end, BM macrophages (BMM) were used as
carriers for nanoparticle-formulated indinavir
(NP-IDV), and the cell distribution was monitored
by single photon emission computed tomography
(SPECT), T
2
* weighted magnetic resonance imag-
ing (MRI), histology, and -scintillation spectrom-
etry. BMM labeled with super paramagnetic iron
oxide and/or
111
indium oxine were infused i.v. into
naı¨ve mice. During the first 7 h, greater than 86%
of cell label was recorded within the lungs. On Days
1, 3, 5, and 7, less than 10% of BMM were in
lungs, and 74 81% and 13–18% were in liver and
spleen, respectively. On a tissue-volume basis, as
determined by SPECT and MRI, BMM densities in
spleen and liver were significantly greater than
other tissues. Migration into the lymph nodes on
Days 1 and 7 accounted for 1.5–2% of the total
BMM. Adoptive transfer of BMM loaded with NP-
IDV produced drug levels in lymphoid and nonlym-
phoid tissues, which exceeded reported therapeu-
tic concentrations by 200- to 350-fold on Day 1
and remained in excess of 100- to 300-fold on Day
14. These data show real-time kinetics and desti-
nations of macrophage trafficking and demonstrate
the feasibility of monitoring macrophage-based,
nanoformulated ART. J. Leukoc. Biol. 80:
000 000; 2006.
Key Words: monocytes cell trafficking mouse indinavir
INTRODUCTION
Peripheral blood mononuclear phagocytes (MP), bone marrow
(BM) and blood monocytes, tissue macrophages, microglia, and
dendritic cells (DC) serve as vehicles for dissemination and
reservoirs for the HIV-1 infection [1–3]. The ability of BM-
derived macrophages (BMM) to migrate to the sites of infection
makes them attractive candidates for use as vehicles to deliver
antiretroviral agents. Indeed, the tissue distribution of circu-
lating monocytes and notably, subpopulations of monocytes
(CD14/CD16), which emerge during inflammatory conditions,
closely parallels sites of active viral replication [4, 5].
The question we asked is whether the same MP, which
disseminate virus, could be used to traffic antiretroviral drugs
to tissue sites of active viral replication. Precedent is provided
by cell-based systems already operative for DNA vaccines,
viral gene, and drug delivery systems developed for a variety of
degenerative and neoplastic diseases [6 –9]. However, obsta-
cles in realizing this goal center on drug cell uptake and
accurate real-time longitudinal study of monocyte trafficking to
tissues known to harbor HIV-1 [10]. The first was demonstrated
in our laboratories in cell culture experiments [11]. The second
involves cross-sectional studies of macrophage trafficking and
cell distribution kinetics, which are limited in number [10, 12].
Over 20 years ago, one study, combining radioactive tracer and
histological examinations, demonstrated solely that 45% of
splenic macrophages are locally produced, and 55% are de-
rived from monocytic extravasation [13]. Moreover, a single,
“theoretical “report of mathematical models for the path of
macrophages and their potential as vehicles for drug delivery
was reported without biologic support [14]. Thus, we strove to
use two sensitive methods, magnetic resonance imaging (MRI)
and single photon emission computer tomography (SPECT), to
examine BMM trafficking to test the feasibility of macrophage-
based drug delivery systems. Demonstrable numbers of BMM
in lymphoid tissues by MRI and SPECT analyses compelled us
to examine whether macrophage delivery of nanoformulated
antiretroviral agents could provide rapid and prolonged anti-
retroviral, therapeutic concentrations in tissues, wherein HIV
is harbored and actively replicates.
1
Correspondence: Department of Pharmacology and Experimental Neuro-
science, University of Nebraska Medical Center, 985880 Nebraska Medical
Center, Omaha, NE 68198-5880. E-mail: hegendel@unmc.edu
Received February 21, 2006; revised June 19, 2006; accepted June 20,
2006; doi: 10.1189/jlb.0206110.
0741-5400/06/0080-0001 © Society for Leukocyte Biology Journal of Leukocyte Biology Volume 80, November 2006 1
Uncorrected Version. Published on August 14, 2006 as DOI:10.1189/jlb.0206110
Copyright 2006 by The Society for Leukocyte Biology.
MATERIALS AND METHODS
Animals
Male BALB/c mice (Charles River Laboratory, Inc., Wilmington, MA), 5– 8
weeks old, were used for all experiments. Animals were housed in sterile
microisolator cages and maintained in accordance with ethical guidelines for
the care of laboratory animals of University of Nebraska Medical Center
(Omaha) and National Institutes of Health (NIH; Bethesda, MD).
Preparation of BMM
To obtain BM-derived mononuclear cells, femurs were excised and flushed
with HBSS. RBC were lysed with ammonium chloride potassium lysis buffer,
and clumps were removed by passing the cell suspension through a 40-m cell
strainer. BM cells were cultured for 10 days in Teflon flasks [15] at 2 10
6
cells/ml in DMEM supplemented with 10% FCS, 2 mM L-glutamine, 1%
penicillin/streptomycin, and 2 g/ml M-CSF (complete medium). All reagents
for cell culture were obtained from Invitrogen (Carlsbad, CA), except for
M-CSF, which was a generous gift from Wyeth, Inc. (Cambridge, MA). Half of
the medium was exchanged for fresh medium every 2 days. The purity of
monocytes from culture was determined by flow cytometry using PE-conjugated
antibody to CD11b (BD PharMingen, San Diego, CA) and analyzed with a
FACSCalibur (BD Biosciences, San Jose, CA). CD11b
cells within the BMM
cultures were always detected in excess of 98%.
Magnetic resonance tracking of Feridex-labeled
macrophages
BMM were labeled with super paramagnetic iron oxide (SPIO) particles (Feri-
dex, Berlex Inc., Wayne, NJ) by culturing at 2.5 mg Feridex/10
7
cells/ml
complete media for1hat37°C. Cells were washed twice with DMEM, and
each recipient mouse received 1 10
7
SPIO-labeled BMM in 200 l i.v. via
the tail vein. Greater than 95% cells were labeled with SPIO particles as
determined by enumeration of Prussian blue-stained cells from cytological
preparations. The presence of SPIO-labeled BMM in tissues was evaluated by
MRI. Accumulation of SPIO particles in tissue causes an increase in the
magnetic relaxivity of tissue water, which is strongly field-dependent. Mea-
sures of relaxivity in our laboratories using a 7T system (Bruker 21 cm Biospec
operating Paravision 3.0.2) demonstrated that relaxivity is related directly to
cell density, assuming uniform labeling and no cell death leading to loss of
SPIO to native macrophage activity. This approach was used to track the
migration of peripheral monocytes within the liver and spleen after i.v. injec-
tion [16, 17]. High-resolution, three-dimensional (3D) gradient-recalled echo
MRI scans of mouse body were acquired using a 25-mm birdcage volume coil
covering a region from the neck to the hips with acquisition parameters of TE
3 ms, TR 50 ms, 30% echo, flip angle 35 degrees, NA 2, field of
view 35 25 50 mm with a resolution of 256 128 128 (voxel
size137195390 m), reconstructed to 256 256 128, total acqui-
sition time 30 min. Signal intensity was normalized to an external standard
to account for signal drift over time. BMM accumulation was determined by
signal loss over time within selected regions of interest. After the injection of
SPIO-labeled cells, 3D gradient-recalled echo images were acquired every 30
min for 6.5 h, at 24 h, and on Days 3, 5, and 7 thereafter. Signal intensity,
normalized to an external standard, was measured within anatomical regions of
interest to determine the rate of labeled cell influx or efflux. R
2
* (1/T
2
*)
decreases in direct proportion to the density of labeled cells in the tissue. We
used this to calculate the average cell density from normalized signal loss using
the equation: Cell density ln(M
unlabeled
) – ln(M
SPIO
), whereby cell density is
proportional to the normalized signal intensity before injection of SPIO-labeled
cells (M
unlabeled
) minus the normalized signal intensity at each time-point after
injection (M
SPIO
).
Monocyte tracking using SPECT
To assess cell migration by SPECT, BMM were labeled with 111indium (
111
In)
oxyquinoline (Indium oxine, Amersham Healthcare, Arlington Heights, IL) at
a dose of 600 Ci per 30 106 cells in 1 ml RPMI 1640/10 mM HEPES for
45 min at 37°C. Cells were washed extensively and resuspended in HBSS.
Labeling efficiency, as determined by -scintillation spectrometry (Packard
Instrument Co., Meriden, CT), was routinely 70 80% of total input isotope.
Each recipient received 5–10 10
6 111
In-labeled BMM i.v. Mice were
anesthetized with 0.5–1% isoflurane delivered in a 2:1 mixture of nitrous oxide
and oxygen. Image acquisition was accomplished with a -scintillation camera
detector fitted with a 1-mm pinhole collimator and interfaced with image
acquisition software (A-SPECT, Gamma Medica, Northridge, CA). Briefly for
each animal, 64 1-min, equiangular exposures over a 360° axis of rotation were
acquired at each time-point. Acquired exposures were reconstructed into a
single 3D tomogram. Regions of interest (ROI) within the processed tomograms
were circumscribed by electronic bit maps to contain the lungs, liver, or
spleen, and relative activities for each were determined. After acquisition of
SPECT images for final time-point, animals were killed, and tissues excised,
weighed, and submitted for -scintillation spectrometry to determine the
intensity of
111
In signal in each tissue. After scintillation spectrometry, tissue
was processed for autoradiography. Frozen sections of 30 m were obtained
and exposed to X-ray film. Autoradiographic images were digitized, and
intensities of
111
In-labeled BMM were assessed by digital image analysis
(MCID Image Analysis System, Imaging Research, Inc., St. Catherines, On-
tario, Canada).
Histology
Tissues were collected from mice after MRI tracking, fixed in 4% parafarm-
aldehyde for 24 h, and embedded in paraffin, and 5 m-thick sections were cut
for analysis. Slide-mounted sections were deparaffinized, rehydrated, and
reacted for 30 min in 2% potassium ferrocyanide in 3.7% hydrochloric acid to
visualize ferric iron particles by Prussian blue. Stained sections were washed
and counter-stained with nuclear fast red. Replicate serial sections were
stained with H&E to provide histological cell distributions. Images of stained
sections were imported into Image-Pro Plus, Version 4.0 (Media Cybernetics,
Silver Spring, MD) for quantifying Prussian blue-stained cells. As a control for
tissue distributions obtained by cell-free Feridex, Feridex alone was adminis-
tered i.v. to naı¨ve recipients at 0.25 mg for each mouse, equivalent to the
amount used to load 10 10
6
. Spleen, liver, lung, and lymph nodes were
collected on Eay 5 after Feridex administration, paraffin-embedded, sectioned,
and stained for Prussian blue.
Indinavir (IDV) loading of BMM, adoptive transfer,
and detection in tissues
BMM were incubated with 5 10
4
M nanoparticle-formulated IDV (NP-IDV;
Baxter Healthcare Corp., Round Lake, IL) in complete medium for 12 h of
incubation at 37°C. Cells were washed and adoptively transferred into recip-
ient mice via tail vein. Animals were killed on Days 1 and 14, and tissues were
collected for analysis of IDV concentrations by reverse phase (RP)-HPLS, as
modified by the method of Jayewardene et al. [18]. Briefly, tissues were
homogenized in 60% methanol, maintained at 4°C overnight, and clarified by
centrifugation at 20,000 g for 15 min at 4°C. Supernatants were collected and
added to glass tubes containing 1 ml diethyl ether. The tubes were mixed for
30 s and maintained at –20°C for 30 min. The ether layer was evaporated to
dryness under a nitrogen stream at room temperature-to-dryness. The residue
was reconstituted in 150 L mobile phase [10 mM ammonium dihydrogen
phosphate with 1 mM 1-heptanesulfonic acid at pH 4.8 mixed with acetonitrile
65:35 (v/v)]. The rehydrated samples were clarified by centrifugation at 20,000
g for 5 min. Triplicate 35 l aliquots of each sample were injected for
RP-HPLC analysis (Shimadzu Corp., Columbia, MD). A C
4
RP column with 5
m particle size packing (Phenomenex, Torrance, CA) was used, and analytes
were measured at 210 nm. The data were analyzed using chromatographic
software (EZStart, Shimadzu Corp.), and peak area was integrated, concentra-
tions of IDV were determined compared with a standard concentration curve of
IDV in mobile phase. Processing and analyses were validated by spiking
known concentrations of IDV in homogenized tissue samples from naı¨ve
animals.
NP-IDV was tagged with Lissamine™ rhodamine B 1,2-dihexadecanoyl-sn-
glycero-3-phosphoethanolamine, triethylammonium salt (rDHPE; Invitrogen).
BMM were loaded with rhodamine-labeled NP-IDV (rh-NP-IDV) and adop-
tively transferred i.v. to naı¨ve recipient mice. Frozen sections of lymphoid and
nonlymphoid tissues were reacted with anti-CD11b (Serotec, Raleigh, NC) and
Alexa Fluor 488 (Molecular Probes, Eugene, OR)-labeled secondary antibody
and evaluated by fluorescence microscopy to detect rh-NP-IDV (red) and
CD11b
(green) cells.
2 Journal of Leukocyte Biology Volume 80, November 2006 http://www.jleukbio.org
Statistical analyses
Normative data from SPECT, MRI, -scintillation spectrometry, and counts
from histology were evaluated by Student’s t-test and ANOVA with least
significant difference post-hoc test using the statistical software packages,
SPSS, v13.0 (SPSS, Inc., Chicago, IL) and Statistica, v7.1 (StatSoft, Inc., Tulsa,
OK). Data are expressed as means SEM for 4 6 animals per group.
Significance levels were chosen as P 0.05.
RESULTS
Tracking
111
In-labeled BMM migration by SPECT
To assess real-time migration of macrophages, 5–7 10
6
111
In-labeled BMM were adoptively transferred to naı¨ve recip-
ients, and migration was assessed by SPECT between 5 and 8 h
post-transfer and on 1, 3, 5, and 7 days thereafter. Within 5 h
post-transfer (Day 0), the majority of radiolabeled BMM was
detected on tomographic images within the lungs with lower
intensity signals emanating from the spleen and liver (Fig.
1A). By Day 1 post-transfer and times thereafter, the most
intense signals were found within the spleen and liver. Con-
gruous signal intensities were demonstrated by all animals.
The progressively decreasing signal intensities exhibited on
Days 3, 5, and 7 reflect the decay characteristics of
111
In
(t
1/2
2.8 days). To quantify the density of BMM within each
tissue, ROI were drawn with electronic bit maps, and the
relative counts and volumes were determined and corrected for
radioactive decay. In agreement with tomographic images, the
number of counts/cm
3
in lungs was significantly greater at 5– 8
h post-transfer compared with those in spleen and liver; how-
ever, by Day 1 and thereafter, counts decreased in lungs and
were increased significantly in spleen and liver, but no signif-
icant differences in densities of BMM were discernible be-
tween liver and spleen (Fig. 1B). Evaluation of percent distri-
bution of BMM indicated that after 5 h post-transfer, 66%
2% of the labeled BMM were in the lung, 25% 2% in the
liver, and 9% 1% in the spleen (Fig. 1C). Later, significantly
greater levels of BMM (74 81%) remained in the liver, 13–
17% in spleen, and 6 –9% in the lungs.
Tracking Feridex-labeled BMM by MRI and
coregistration with SPECT
To evaluate BMM migration by MRI, Feridex-loaded BMM
were adoptively transferred i.v. to naı¨ve recipients, and migra-
tion was tracked over a 7-day period. BMM density exponen-
tially increased in the spleen (Fig. 2A) at a rate of 6.4 h
compared with 8.7 h for liver (Fig. 2, B and C). Cell density in
liver and spleen was significantly higher by Day 1 after transfer
compared with MRI signals at initiation of image acquisition
(time0) and remained significantly higher throughout the
study. A higher but insignificant density of cells was demon-
strated in the spleen compared with liver; however, the relative
concentration of cells within the spleen on Day 7 appears to be
underestimated in MRI compared with SPECT, histology, and
tissue scintillation measures. As a control for migration into a
highly vascular but nonlymphoid tissue, kidneys were evalu-
ated and showed negligible signals, especially after Day 1, and
only a slight increase in BMM density was detected over the
7-day time-course, a finding also appreciated in SPECT im-
ages, indicating little BMM migration into the kidneys. Tissue
movement during vital examination precluded analysis by MRI
in the lungs of viable animals.
To validate MRI and SPECT analyses, BMM were dual-
labeled with Feridex and
111
In and extensively washed, and
10 10
6
were adoptively transferred to recipient mice. Re-
constructed SPECT images were coregistered to MRI scans
using the Analyze package (AnalyzeDirect, Inc.). SPECT data
were interpolated to the resolution of the MRI data
(256256128), and fiducial markers were used for scaling
and alignment. Regions of interest were drawn on MRI and
transferred to interpolated SPECT images to obtain region
counts in fiducial markers and signal intensity in T
2
* weighted
MRI. Coregistered images showed the existence of dual-la-
beled BMM in spleen and liver with increased intensities in
SPECT images (green) as a result of
111
In label, which corre-
sponded to Feridex-induced loss of signal in MRI images (red;
Fig. 2D and Supplemental Video 1). Light-green intensities
around the areas of kidney demonstrated that although signal
loss is negligible by MRI, migration of BMM to kidney is
detectable by SPECT. Coregistration with strong signal-to-
noise ratios at optimal operating efficiencies provided strong
correlation between MRI and SPECT data for spleen (r0.66)
and liver (r0.77). These data confirmed the separate MRI and
SPECT analyses preformed previously (Figs. 1 and 2, A–C),
which indicated that the majority of BMM migrated to spleen
and liver after Day 1 post-transfer and was retained in those
tissues at times thereafter.
Tissue distribution of
111
In-labeled BMM
To validate SPECT analysis, we assessed tissue distribution
and density of
111
In-labeled macrophages by -scintillation
spectrometry of lymphoid (spleen and lymph nodes) and non-
lymphoid (liver, lung, and kidney) tissues harvested immedi-
ately after the last SPECT data acquisition on Days 1 and 7.
After i.v. administration of BMM, spleen, liver, and lung re-
tained 96% of the tissue counts (62.5, 21.3, and 12.1%,
respectively) after Day 1 and were essentially unchanged by
Day 7 (68.2, 20.6, and 6.7%, respectively). Rank order anal-
ysis of BMM density (relative counts/mg tissue weight) and
distribution of counts from spleen and liver indicated signifi-
cant differences (P0.002) between those two tissues and from
other tissues; however, no significant differences were detected
among other tissues. As the densities and tissue distributions
of BMM were similar on Days 1 and 7 post-transfer, this
suggested that after initial migration into tissues, the bulk of
macrophages is retained in those tissues. On Days 1 and 7,
distribution of BMM in lymph nodes (brachial, cervical, and
inguinal) accounted for 2% 1.6% and 1.5% 0.7%, re-
spectively, of the total radioactive signal, suggesting that mac-
rophages possess the capacity to migrate to and remain within
lymph nodes. Despite negligible signals in MRI tracking,
-scintillation spectrometry indicated that kidneys retained
1.9% 1.4% of total
111
In-labeled BMM measured in tissues,
demonstrating that BMM are capable of migrating through
nonlymphoid tissues as well.
Gorantla et al. Macrophage migration and delivery of antiretroviral drugs 3
Days
07531
Lg
Sp
Lv
Lv
Sp
Lv
B
A
C
Sp
Sp
Sp
Lv
Lv
Lv
Lv
Lv Lv
Fig. 1. SPECT analysis to track BMM after adoptive transfer to naı¨ve mice. Mouse BMM were labeled with
111
In oxine [half-life (t
1/2
)2.8 days] and washed,
and 1 10
7
were adoptively transferred by i.v. injection to naı¨ve, syngeneic recipients. Trafficking of BMM was evaluated by SPECT analysis for each animal
after 6 h (Day 0) and 1, 3, 5, and 7 days post-transfer. (A) SPECT images of all four mice on Days 0, 1, 3, 5, and 7, showing congruous intensity of
111
In-labeled
BMM in lung (Lg), liver (Lv), and spleen (Sp). (B) For each lung, liver, and spleen within a tomographic image, a ROI was circumscribed by electronic bit maps
to include each tissue and saved as a subimage. The tomographic subimage was sectioned transaxially, and the relative number of counts and volumes was
determined by image analysis software provided by the manufacturer. Relative counts were corrected for decay, and the mean counts/cm
3
SEM (n4) for each
tissue are plotted as a function of time. (D) The distribution of labeled BMM was determined by normalizing the total relative counts as a percentage of total counts
within all tissues for each animal, and the mean distribution
SEM (n4) was evaluated as a function of time after transfer of BMM.
a
, P 0.05, for spleen and
liver compared with lung.
4 Journal of Leukocyte Biology Volume 80, November 2006 http://www.jleukbio.org
Histological confirmations of Feridex-labeled
BMM
To validate migration of Feridex-labeled macrophages after
adoptive transfer, tissues were harvested from recipient mice
after the last MRI examination at 24 h and from another set
after the last MRI examination on Day 7 post-transfer. Sections
from lung, liver, spleen, and lymph node were processed and
stained to detect Feridex-labeled BMM as Prussian blue-
stained cells. Tissues from uninjected control animals without
Feridex were stained for Prussian blue to detect endogenous
ferrum (data not shown), and this baseline stain was used for
comparison and analysis. Histological examination of tissues
following acquisition of MRI images confirmed that the major-
ity of macrophages migrated to spleen, liver, lung, (Fig. 3), and
lymph nodes (Fig. 4). When quantified using Image-Pro soft-
ware, spleens showed the greatest density of Prussian blue-
stained cells on Days 1 and 7 without detectable, significant
differences between days (55525 and 47335 Prussian blue
cells/mm
2
, respectively; P0.07). Prussian blue-stained BMM
were distributed primarily within the resident macrophage
areas surrounding the germinal centers (Fig. 3). Lungs showed
an initial accumulation of Feridex-labeled cells by 24 h post-
transfer (23719 cells/mm
2
), but virtually no cells could be
detected by Day 7 (1.10.3 cells/mm
2
), demonstrating a sig-
nificant reduction in BMM in the lung over the final 6 days
after transfer (P0.0001). Feridex-labeled cells were detected
in the sinusoids of the liver. The density of BMM in the liver
on Day 1 (819 cells/mm
2
) was significantly less than densi-
ties in the spleen and lung at the same time (P0.0001) but
significantly increased by Day 7 (12110 cells/mm
2
,
P0.009) exceeding levels in lung (P0.001), although re-
mained less than BMM density in spleen at that time
(P0.0001).
Feridex-labeled cells were detected in the lymph nodes from
animals on Day 1 after cell transfer within afferent lymphatics.
Increased magnifications (Fig. 4, C and D) of cortical and
paracortical areas of a representative lymph node (Fig. 4A)
show Feridex-labeled BMM in the interfollicular regions. Mac-
rophages, which are lightly stained for Prussian blue (Fig. 4D),
may represent macrophages with less Feridex as a result of
partial expulsion or death upon reaching the lymph nodes. By
Day 7, significantly higher amounts of Feridex-loaded macro-
phages (19.53.5 cells/mm
2
, P0.004) were detected in the
lymph nodes (Fig. 4, E, G, and H), indicating that macrophages
were capable of reaching lymph nodes, tissues that the drug
must reach for the greatest efficacy against HIV-1 infection and
reservoirs.
Animals were also injected with Feridex alone and tissues
collected on Day 5. When stained for Prussian blue, tissues
from mice administered Feridex alone showed uniquely differ-
Spl
Liv
100%
0%
013
5
Days
B
C
D
A
Spl
Spl Spl
Liv
Liv
Liv
Kid
Bwl
Fig. 2. Tracking BMM migration by MRI and coreg-
istration with SPECT images. (A) Time series of spleen
(Spl), kidneys (Kid), and bowel (Bwl; upper panels) and
liver (Liv; lower panels) from mice before (Day 0) and
on 1, 3, and 5 days after i.v. injection of SPIO (Feri-
dex)-labeled BMM, demonstrating visible signal loss
after injection of Feridex-labeled BMM. Quantitation of
in vivo tracking of mouse BMM labeled with Feridex
using T
2
* weighted MRI during (B) the initial 6.5 h
after adoptive transfer and (C) on Days 1, 3, 5, and 7
thereafter. Results shown are measures of Feridex-
labeled cell densities in spleen, liver, and kidney
(mean
SEM from n4 mice per group).
a
, P 0.05,
compared with kidney. (D) BMM were dual-labeled
with Feridex and
111
In and transferred i.v. to recipient
mice. Recipients were anesthetized and positioned in
custom-built, MRI/SPECT-compatible holders with at-
tached fiducial markers (not shown), and serial acqui-
sitions of MRI and SPECT scans were performed on
Day 1 after transfer. SPECT data were interpolated to
the resolution of the MRI data (256256128), and
fiducial markers were used for scaling and alignment.
Reconstructed SPECT images (green) were coregis-
tered and overlaid to MRI scans (red) using the Analyze
package (AnalyzeDirect, Inc., Lenexa, KS). Displayed,
coregistered images are 390 m slices containing
spleen and kidney (left panel) and liver (right panel)
and show areas of radioactive intensities of
111
In-la-
beled BMM in SPECT images (green), which corre-
spond to loss of MRI signal as a result of Feridex-
labeled BMM.
Gorantla et al. Macrophage migration and delivery of antiretroviral drugs 5
ent patterns of Feridex distribution compared with tissues from
mice administered Feridex-labeled BMM (Fig. 5). Livers from
mice injected with Feridex showed higher accumulations of
Prussian blue-staining cells than those injected with Feridex-
loaded BMM. Spleens showed robust staining only from ani-
mals treated with Feridex-labeled BMM.
Tissue distribution of dual-labeled BMM
To validate measurements of BMM tissue distribution, we
evaluated tissues from mice treated with BMM, which were
dual-labeled with Feridex and
111
In. Spleen, liver, lungs, and
lymph nodes were excised from mice on Day 5 after treatment
with dual-labeled BMM. Tissues were fixed, paraffin-embed-
ded, sectioned, and subjected to autoradiography and histolog-
ical analysis. Digital image analysis (Fig. 6A) and quantifica-
tion (Fig. 6B) of the autoradiographic images show that spleen
retained the highest amount of radioactivity compared with
liver and lung. In spleen, the highest intensity signals were
observed to be concentrated in macrophage-rich areas sur-
rounding the germinal centers. Measurement of radioactivity in
lymph nodes by -scintillation spectrometry confirmed the
migration and presence of
111
In-labeled BMM in lymph nodes
(Fig. 6C).
Uptake and distribution of BMM NP-IDV
IDV was readily taken up by BMM as evident by the intense
black NP inclusions visualized by light microscopy in BMM
cocultured in the presence of NPs (Fig. 7B) compared with
those cultured in the absence of NPs (Fig. 7A). Virtually 100%
of the BMM retained the NPs. RP-HPLC analysis of BMM
lysates revealed an IDV concentration of 37.5 M or 75% of
the initial NP-IND concentration [21]. To evaluate the poten-
tial for delivery of NP-IDV by macrophages and assess whether
therapeutic concentrations of IDV can be attained in immune
tissues, we loaded BMM with NP-IDV, as shown in Figure 7B,
and adoptively transferred the NP-IDV-loaded BMM i.v. to
naı¨ve, recipient mice. IDV concentrations were evaluated by
RP-HPLC in spleen, lymph nodes, lung, liver, and kidney on
Days 1 and 14 after transfer (Fig. 7C). Of foremost significance
is the finding that for all tissues examined, IDV concentrations,
on a tissue-weight basis, were at least one order of magnitude
in excess of the reported therapeutic concentration range in
human plasma [19, 20] and were at least 2 and 3 orders of
magnitude higher, respectively, in the lymph nodes and spleen;
both lymphoid tissues implicated in harboring HIV-1 reser-
voirs. One day after transfer of NP-IDV-loaded BMM, IDV
concentrations were significantly highest in the lung compared
with other tissues, reaching levels that were in excess of 3.5
orders of magnitude greater than the therapeutic concentration
range of IDV in plasma. It is notable that significant levels of
IDV were detected in spleen and lymph nodes. By Day 14 after
transfer, IDV concentrations were significantly higher in lym-
phoid tissues of the spleen and lymph nodes compared with
other nonlymphoid tissues of lung, liver, and kidney. Although
IDV concentrations were not changed significantly in lymph
nodes between Days 1 and 14, levels increased significantly in
spleen by Day 14 and diminished in lungs, liver, and kidney.
DISCUSSION
Macrophages have been of great interest as gene and drug
delivery vehicles [9, 22]. However, relatively few in vivo stud-
ies have assessed the ability of ex vivo-transferred macro-
phages to migrate to the target sites. For optimal use of mac-
rophage delivery systems, such studies are necessary to accu-
rately assess whether cells and drugs reach a targeted
distribution. In this study, we explored the feasibility of using
macrophages for delivery of nanoformulations of the anti-
HIV-1 protease, IDV, with the expressed intent to improve the
efficacy of the drug by targeted distribution. A necessary step
toward implementing this strategy required the measurement of
rh-NP-IDV
CD11b
Prussian
Blue
Spleen
H & E
LungLiver
Germinal center
Interfollicular area
50
50
µm
5 µm
Fig. 3. Feridex or NP-IDV-loaded macrophages within
lymphoid and nonlymphoid tissues after transfer to naı¨ve
recipients. BMM were loaded with Feridex or rhodamine-
labeled NP-IDV (rh-NP-IDV) and injected i.v. into naı¨ve
recipients. Spleen, lung, and liver were harvested and
evaluated by immunohistological analysis. After acquisi-
tion of final MRI images after Day 1 post-transfer, tissues
were stained to detect Feridex-labeled macrophages by
Prussian blue staining (blue) and counter-stained with nu-
clear fast red. Serial sections were stained with H&E for
histological comparison. Feridex-labeled BMM were de-
tected in areas of resident interfollicular macrophages sur-
rounding the germinal centers. Tissues from mice receiving
rh-NP-IDV-loaded BMM were collected at Day 5 after
adaptive transfer, processed as frozen sections for immu-
nohistology, and examined by fluorescent microscopic
analysis. CD11b (green)-positive macrophages containing
rhodamine-labeled NPs (red) are shown in spleen, liver,
and lung. Original, large micrograph magnifications, 200
m; original bar, 50 m; original inset magnifications,
1000; bar, 5 m.
6 Journal of Leukocyte Biology Volume 80, November 2006 http://www.jleukbio.org
macrophage migration and homing to ensure the adequacy of
cellular penetration into tissues wherein HIV is harbored and
replicates.
For these studies, we used syngeneic BMM as sources
originating from normal progenitor cells, which were expanded
by in vitro cultivation. For tracking studies, we used BMM as
radiolabeled (
111
In) macrophages or macrophages laden with
Feridex. The former were tracked by SPECT and the latter by
T
2
* MRI. Labeled BMM were adoptively transferred i.v. to
naı¨ve recipient mice, and recipients were subjected to MRI
and SPECT analyses. Images were initially obtained every
30 60 min during the first 8 h post-transfer and then, 24 h and
every other day thereafter. Clearly, as assessed by MRI and
SPECT, the majority of BMM initially remained in the lung but
infiltrated to the spleen by 24 h and remained within lymphoid
tissue for up to 7 days. -Scintillation spectrometry and his-
tological examination of spleen confirmed the presence of
labeled BMM and further demonstrated their presence in
lymph nodes. Although the majority of BMM was retained in
the liver as a result of the mass of the tissue, the densities of
BMM in spleen and liver on a volume basis were not signifi-
cantly different from each other, as determined by SPECT and
MRI. Several studies reported similar pattern of distribution of
systemically administered macrophages [10, 12]. Migration of
macrophages into the spleen was shown to be dependent on
their activation state. Murine peritoneal macrophages elicited
by Brewer’s thioglycollate medium when injected i.v. localized
in the lung with minimal migration to spleen even after 72 h
[10]. Macrophage-like cell lines were shown to have no migra-
tion capability but were rapidly cleared from the circulation.
SPECT and MRI provide invaluable tools for noninvasive
longitudinal assessment of migration and distribution of cell
trafficking after transfer of leukocytes in animals and humans.
SPECT is a sensitive method to follow the time-course of
migration of transplanted cells using
111
In-oxine-labeled cells
[23].
111
In has been used in studies to find biodistribution of
the cells after transplantation, but animals typically had to be
killed at each time-point for autoradiography or radioactivity
measurement [24]. SPECT scanning allows in vivo real-time
tracking of cell trafficking in the same animal or patient.
111
In-oxine labeling of peripheral blood leukocytes to assess
foci of inflammation has long been used in clinical studies of
inflammatory disorders [25, 26]. Moreover, the use of
111
In-
labeled macrophages to study the distribution of mononuclear
cells in tumors was demonstrated as an elegant procedure for
tracking the distribution of macrophages and DC [27–30].
50 µm 50 µm
Fig. 5. Tissue distributions from mice treated with BMM loaded with Feridex
or Feridex alone. Animals were injected i.v. with 10 10
6
BMM loaded with
0.25 mg Feridex or 0.25 mg Feridex alone. Tissues were obtained on Day 5
postadministration and paraffin-embedded, and sections were subjected to
Prussian blue staining. Feridex was distributed uniquely when injected as a
cell-free formulation compared with BMM-loaded Feridex showing greater
accumulation of Feridex in liver and less in spleen. Original micrograph
magnifications, 100; bar, 50 m.
100 µm
100 µm
100 µm
100 µm
10 µm
10 µm 10 µm
25 µm
Fig, 4. BMM migration to the lymph nodes, which were extracted from mice
at (A–D) Day1 and (E–H) Day 7 after adaptive transfer of Feridex-labeled
BMM. (A, C–E, G, H) Sections were stained to detect Feridex using Prussian
blue staining and counterstained with nuclear fast red. (B, F) Serial sections
were stained with H&E for histological comparison to show the afferent
lymphatics marked by the arrow. (A, B, E, F) Micrographs are 100 original
magnifications. (C, D) Micrographs are 1000 original magnifications of the
areas marked in A showing (C) Feridex-labeled cells and (D) many degener-
ating macrophages faintly labeled with Feridex. Panels are (G) 400 original
magnification and (H) 1000 original magnification of the boxed area shown
in E containing released Feridex and Feridex-labeled cells in the afferent
lymphatics.
Gorantla et al. Macrophage migration and delivery of antiretroviral drugs 7
Similarly, MRI provides another noninvasive method for lon-
gitudinally studying in vivo, the fate of transplanted cells
labeled with a paramagnetic probe such as Feridex [31], there-
fore rendering cell-specific imaging an increasingly important
field of MRI [16, 32–34].
Having shown by SPECT, MRI, -scintillation spectrometry,
and histological examination that adoptive transfer of unlad-
ened and NP-ladened BMM provided detectable and prolonged
levels of macrophage migration into lymphoid tissues, whether
adoptive transfer of macrophages ladened with antiretroviral
agents can be used for efficacious, antiretroviral drug delivery
has not been assessed. Adoptive transfer of macrophages has
been shown to be a relatively risk-free procedure in humans
with only mild side effects [35, 36] and should provide a
unique, efficacious delivery platform by which to introduce
antiretroviral agents deeper into lymphoid tissues. To assess
the efficacies of antiretroviral distribution into tissues where
HIV replicates, strategies were initiated to deliver antiretrovi-
ral drugs in macrophage vehicles, cells that naturally migrate
to tissues associated with HIV infection and harboring HIV
reservoirs. Our data demonstrated that with one administration
of macrophages loaded with NP-IDV, therapeutic levels of IDV
in lymphoid and nonlymphoid tissues were attained rapidly by
1-day postadministration and were maintained in the spleen,
lung, and lymph nodes for up to 14 days thereafter. These
observations are supported by investigations in humanized
mice infected with HIV-1. Here, NP-IDV administered in
BMM demonstrates antiretroviral responses and protection of
human CD4 T cells [21]. It is interesting that although IDV
levels were highest in the lung on Day 1 compared with other
tissues, BMM in the lung were relatively low as determined by
SPECT for
111
In-labeled BMM and by MRI using Feridex-
labeled BMM. This suggested that by Day 1 after transfer, a
sufficient number of NP-IDV-loaded BMM remained in the
lung to release such high levels of IDV or high levels of
NP-IDV or free IDV remained in the lung after cells had
immigrated, possibly as a result of release from BMM and
subsequent uptake by endogenous lung macrophages. The
kinetics and biodistribution of free IDV given orally have been
shown to be short-lived and require multiple doses per day to
attain and maintain therapeutic levels [37]; nanoformulated
IDV delivered via macrophage vehicles afforded rapid and
prolonged attainment of therapeutic levels of IDV in tissues
where HIV is harbored and replicates. Although the levels
reported here resulted from a single administration of NP-IDV-
loaded IDV, the potential for higher concentrations remains
possible upon multiple doses. Therefore, these findings
strongly support the feasibility of using macrophages as drug
Germinal
center
100 µm 100 µm100 µm
Fig. 6. Distribution of
111
In/Feridex-labeled BMM,
which were dual-labeled with Feridex and
111
In, and 10
10
6
-labeled cells were adoptively transferred to recipient
mice i.v. Tissues were collected on Day 5 after BMM
administration, and 30 m frozen sections were obtained
and exposed to X-ray film for autoradiography. Autoradio-
graphic images were digitized and assessed by digital
image analysis. (A) Original autoradioraphic images (top
panels) and enlarged areas (center panels) show radioac-
tive signal intensities from
111
In-labeled BMM in spleen,
liver, and lung. Higher radioactive signal is observed in
splenic interfollicular areas around germinal centers. The
same sections used for autoradiography were visualized for
Feridex by Prussian blue staining (bottom panels) and
nuclear fast red counterstain. A splenic germinal center is
depicted (circle) surrounded by Prussian blue-stained
BMM in interfollicular areas (arrows). Original magnifica-
tions for bottom panels are 100. (B) Intensities of radio-
active signals in tissues from the images collected from six
animals were quantified and expressed as mean
SEM. (C)
Radioactivity in lymph nodes collected from animals on
Day 5 after BMM administration was measured by -scin-
tillation spectrometry and graphed as cpm/mg tissue
weight (meanSEM of six animals). In LN, Inguinal lymph
node; Br LN, brachial lymph node; Cer LN, cervical lymph
node.
8 Journal of Leukocyte Biology Volume 80, November 2006 http://www.jleukbio.org
delivery vehicles for nanoformulated drugs to enhance the
efficacy of antiretroviral therapeutics. This strategy has been
shown to reduce markers of infectious HIV-1 in serum and
lymphoid tissues of a human/mouse chimera model of HIV-1
infection [21].
Antiretroviral therapy (ART), which includes IDV as a po-
tent protease inhibitor, has been effective in reducing plasma
viral load in HIV-infected patients. The efficacy of ART is
compromised typically as a result of the nature of complex
regimens requiring multiple daily dosing, diligent adherence to
the regimen, and the limited biodistribution of an orally ad-
ministered drug [38]. The efficacy of IDV use is also compro-
mised, as administration of an unformulated drug results in low
levels in deep lymphoid organs [39], where HIV infection is
prevalent as a result of trapped HIV particles on the follicular
DC in the germinal centers [40 42]. Viral replication in
lymphoid tissues has been shown to be ten- to100-fold higher
than that in PBMC [41]. Using higher doses of antiretroviral
agents can increase the drug concentration within lymphoid
tissues; however, the drawback is that this strategy for attain-
ment of therapeutically effective concentrations produces toxic
side effects by many antiretroviral drugs and often complicates
HIV drug therapies [38, 43]. Such adverse effects could be
minimized by nanoformulated drug delivery modalities that
result in selective uptake of drug by macrophages [9, 44, 45]
and provide site-specific delivery to the lymphoid tissues, thus
preventing HIV replication and the establishment of HIV
reservoirs during a clinical latency period.
ACKNOWLEDGMENTS
This work was supported by NIH Grants P01 NS31492, R01
NS34239, R37 NS36136, and P01 MH64570 (to H. E. G.) and
P01 NS43985 (to M. B. and H. E. G.). The authors thank Robin
Taylor of the University of Nebraska Medical Center for ad-
ministrative assistance.
REFERENCES
1. Gendelman, H. E., Meltzer, M. S. (1989) Mononuclear phagocytes and the
human immunodeficiency virus. Curr. Opin. Immunol. 2, 414 419.
2. Verani, A., Gras, G., Pancino, G. (2005) Macrophages and HIV-1: dan-
gerous liaisons. Mol. Immunol. 42, 195–212.
3. Kedzierska, K., Crowe, S. M. (2002) The role of monocytes and macro-
phages in the pathogenesis of HIV-1 infection. Curr. Med. Chem. 9,
1893–1903.
4. Crowe, S., Zhu, T., Muller, W. A. (2003) The contribution of monocyte
infection and trafficking to viral persistence, and maintenance of the viral
reservoir in HIV infection. J. Leukoc. Biol. 74, 635– 641.
5. Scherberich, J. E. (2003) Proinflammatory blood monocytes: main effector
and target cells in systemic and renal disease; background and therapeutic
implications. Int. J. Clin. Pharmacol. Ther. 41, 459 464.
6. Griffiths, L., Binley, K., Iqball, S., Kan, O., Maxwell, P., Ratcliffe, P.,
Lewis, C., Harris, A., Kingsman, S., Naylor, S. (2000) The macrophage—a
novel system to deliver gene therapy to pathological hypoxia. Gene Ther.
7, 255–262.
7. Tanaka, S., Kitagawa, K., Sugiura, S., Matsuoka-Omura, E., Sasaki, T.,
Yagita, Y., Hori, M. (2004) Infiltrating macrophages as in vivo targets for
intravenous gene delivery in cerebral infarction. Stroke 35, 1968 –1973.
8. Verma, I., Pandey, R., Khuller, G. K. (2004) Liposomes as adjuvant for
anti-mycobacterial vaccine development. Indian J. Exp. Biol. 42, 949
954.
9. Basu, M. K., Lala, S. (2004) Macrophage-specific drug delivery in exper-
imental leishmaniasis. Curr. Mol. Med. 4, 681– 689.
10. Wiltrout, R. H., Brunda, M. J., Gorelik, E., Peterson, E. S., Dunn, J. J.,
Leonhardt, J., Varesio, L., Reynolds, C. W., Holden, H. T. (1983) Distri-
bution of peritoneal macrophage populations after intravenous injection in
mice: differential effects of eliciting and activating agents. J. Reticuloen-
dothel. Soc. 34, 253–269.
11. Dou, H., Limoges, J., Ellison, B., Kingsley, J. D., Zhou, Y., Destache,
C. J., Chaubal, M., Werling, J., Kipp, J., Rabinow, B., et al. (2006)
Macrophage-based delivery system for antiretroviral therapy.
12. Kennedy, D. W., Abkowitz, J. L. (1998) Mature monocytic cells enter
tissues and engraft. Proc. Natl. Acad. Sci. USA 95, 14944 –14949.
13. Van Furth, R. (1992) Development and Distribution of Mononuclear
Phagocytes, New York, NY, Raven.
14. Owen, M. R., Byrne, H. M., Lewis, C. E. (2004) Mathematical modeling of
the use of macrophages as vehicles for drug delivery to hypoxic tumor
sites. J. Theor. Biol. 226, 377–391.
15. van der Meer, J. W., Bulterman, D., van Zwet, T. L., Elzenga-Claasen, I.,
van Furth, R. (1978) Culture of mononuclear phagocytes on a teflon
surface to prevent adherence. J. Exp. Med. 147, 271–276.
16. Zelivyanskaya, M. L., Nelson, J. A., Poluektova, L., Uberti, M., Mellon,
M., Gendelman, H. E., Boska, M. D. (2003) Tracking superparamagnetic
iron oxide-labeled monocytes in brain by high-field magnetic resonance
imaging. J. Neurosci. Res. 73, 284 –295.
17. Terrier, F., Tourniaire, J., Belenger, J., Slosman, D., Rubbia, L., Grossh-
olz, M., Mentha, G., Hiltbrand, E., Mermillod, B. (1995) Magnetic reso-
nance imaging with superparamagnetic iron oxide particles to evaluate
hepatic macrophage-monocytic phagocytosis after arterial devasculariza-
tion in minipigs. Acad. Radiol. 2, 565–575.
Fig. 7. IDV levels in lymphoid and nonlymphoid tissues after i.v. adminis-
tration of NP-IDV-loaded BMM. Mouse macrophages (BMM) were incubated
for 12 h at 37°C in 5% CO
2
with NP-IDV at a final concentration of 5 10
4
M. Light micrographs of (A) unloaded macrophages and (B) NP-IDV-loaded
macrophages are shown at 400 original magnification. (C) NP-IDV-loaded
BMM were administered i.v. to recipient mice. On Days 1 and 14 postadmin-
istration, tissues were harvested, weighed, homogenized, and submitted for
RP-HPLC analysis for levels of IDV. Means of IDV concentrations in ng/g
tissue wet weight (SEM) of six mice per time-point were determined and
plotted as a log function. Therapeutic concentration range for IDV in human
plasma (150 675 ng/ml plasma) [19, 20] is indicated as the translucent gray
band parallel to the x-axis. Comparison of IDV concentrations between tissues
within each time-point: P 0.05 is significant compared with
a
, spleen;
b
,
lymph nodes;
c
, lung; and
d
, liver.
e
, P 0.05, kidney, is significant for
comparison of IDV concentrations between Day 1 and Day 14 for each
respective tissue.
Gorantla et al. Macrophage migration and delivery of antiretroviral drugs 9
18. Jayewardene, A. L., Zhu, F., Aweeka, F. T., Gambertoglio, J. G. (1998)
Simple high-performance liquid chromatographic determination of the
protease inhibitor indinavir in human plasma. J. Chromatogr. B Biomed.
Sci. Appl. 707, 203–211.
19. Bossi, P., Peytavin, G., Lamotte, C., Calvez, V., Bricaire, F., Costagliola,
D., Katlama, C. (2003) High indinavir plasma concentrations in HIV-
positive patients co-infected with hepatitis B or C virus treated with low
doses of indinavir and ritonavir (400/100 mg twice a day) plus two
nucleoside reverse transcriptase inhibitors. AIDS 17, 1108 –1110.
20. Lamotte, C., Descamps, D., Joly, V., Duval, X., Peytavin, G. (2002)
Validation of an algorithm for indinavir (IDV) dosage adjustment (IADA)
with patient with high IDV trough plasma concentrations (Cmin) to prevent
adverse events (AE) in a easy and potent IDV/ritonavir (RTV) containing
regimen. In International Workshop on Clinical Pharmacology of HIV
Therapy, Washington, DC.
21. Dou, H., Destache, C. J., Morehead, J. R., Mosley, R. L., Boska, M. D.,
Kingsley, J., Gorantla, S., Poluektova, L., Nelson, J. A., Chaubal, M.,
Werling, J., Kipp, J., Rabinow, B., Gendelman, H. E. (2006) Development
of a macrophage nanoparticle system for antiretroviral drug delivery:
pharmacokinetics, immune, and anti-viral responses in a murine model of
HIV-1 disease. Blood, Epub ahead of print.
22. Burke, B. (2003) Macrophages as novel cellular vehicles for gene therapy.
Expert Opin. Biol. Ther. 3, 919 –924.
23. Bennink, R. J., van Montfrans, C., de Jonge, W. J., de Bruin, K., van
Deventer, S. J., te Velde, A. A. (2004) Imaging of intestinal lymphocyte
homing by means of pinhole SPECT in a TNBS colitis mouse model. Nucl.
Med. Biol. 31, 93–101.
24. Yeager, M. P., DeLeo, J. A., Hoopes, P. J., Hartov, A., Hildebrandt, L.,
Hickey, W. F. (2000) Trauma and inflammation modulate lymphocyte
localization in vivo: quantitation of tissue entry and retention using in-
dium-111-labeled lymphocytes. Crit. Care Med. 28, 1477–1482.
25. Ascher, N. L., Ahrenholz, D. H., Simmons, R. L., Weiblen, B., Gomez, L.,
Forstrom, L. A., Frick, M. P., Henke, C., McCullough, J. (1979) Indium
111 autologous tagged leukocytes in the diagnosis of intraperitoneal
sepsis. Arch. Surg. 114, 386 –392.
26. Becker, W., Meller, J. (2001) The role of nuclear medicine in infection and
inflammation. Lancet Infect. Dis. 1, 326 –333.
27. Eggert, A. A., Schreurs, M. W., Boerman, O. C., Oyen, W. J., de Boer,
A. J., Punt, C. J., Figdor, C. G., Adema, G. J. (1999) Biodistribution and
vaccine efficiency of murine dendritic cells are dependent on the route of
administration. Cancer Res. 59, 3340 –3345.
28. Quillien, V., Moisan, A., Lesimple, T., Leberre, C., Toujas, L. (2001)
Biodistribution of 111indium-labeled macrophages infused intravenously
in patients with renal carcinoma. Cancer Immunol. Immunother. 50,
477– 482.
29. Abreo, K., Lieberman, L. M., Moorthy, A. V. (1985) Distribution studies
of 111In-oxine-labeled peritoneal mononuclear cells in tumor-bearing
rats. Int. J. Nucl. Med. Biol. 12, 53–55.
30. Oluwole, S. F., Fawwaz, R. A., Engelstad, K., Wang, T. S., Hardy, M. A.
(1990) Migration patterns of indium-111-labeled dendritic cells in the rat.
Prog. Clin. Biol. Res. 355, 247–256.
31. Pautler, R. G. (2004) Mouse MRI: concepts and applications in physiol-
ogy. Physiology (Bethesda) 19, 168 –175.
32. Bulte, J. W., Ma, L. D., Magin, R. L., Kamman, R. L., Hulstaert, C. E., Go,
K. G., The, T. H., de Leij, L. (1993) Selective MR imaging of labeled
human peripheral blood mononuclear cells by liposome-mediated incor-
poration of dextran-magnetite particles. Magn. Reson. Med. 29, 32–37.
33. Anderson, S. A., Shukaliak-Quandt, J., Jordan, E. K., Arbab, A. S.,
Martin, R., McFarland, H., Frank, J. A. (2004) Magnetic resonance
imaging of labeled T-cells in a mouse model of multiple sclerosis. Ann.
Neurol. 55, 654 659.
34. Anderson, S. A., Glod, J., Arbab, A. S., Noel, M., Ashari, P., Fine, H. A.,
Frank, J. A. (2005) Noninvasive MR imaging of magnetically labeled stem
cells to directly identify neovasculature in a glioma model. Blood 105,
420 425.
35. Faradji, A., Bohbot, A., Schmitt-Goguel, M., Roeslin, N., Dumont, S.,
Wiesel, M. L., Lallot, C., Eber, M., Bartholeyns, J., Poindron, P., et al.
(1991) Phase I trial of intravenous infusion of ex-vivo-activated autologous
blood-derived macrophages in patients with non-small-cell lung cancer:
toxicity and immunomodulatory effects. Cancer Immunol. Immunother.
33, 319 –326.
36. Andreesen, R., Hennemann, B., Krause, S. W. (1998) Adoptive immuno-
therapy of cancer using monocyte-derived macrophages: rationale, current
status, and perspectives. J. Leukoc. Biol. 64, 419 426.
37. Hoetelmans, R. M. (1999) Pharmacology of antiretroviral drugs. Antivir.
Ther. 4, Suppl 3, 29 41.
38. Casau, N. C., Glesby, M. J., Paul, S., Gulick, R. M. (2003) Brief report:
efficacy and treatment-limiting toxicity with the concurrent use of lopina-
vir/ritonavir and a third protease inhibitor in treatment-experienced HIV-
infected patients. J. Acquir. Immune Defic. Syndr. 32, 494 498.
39. Gagne, J. F., Desormeaux, A., Perron, S., Tremblay, M. J., Bergeron, M. G.
(2002) Targeted delivery of indinavir to HIV-1 primary reservoirs with
immunoliposomes. Biochim. Biophys. Acta 1558, 198 –210.
40. Embretson, J., Zupancic, M., Beneke, J., Till, M., Wolinsky, S., Ribas,
J. L., Burke, A., Haase, A. T. (1993) Analysis of human immunodeficiency
virus-infected tissues by amplification and in situ hybridization reveals
latent and permissive infections at single-cell resolution. Proc. Natl. Acad.
Sci. USA 90, 357–361.
41. Cohen, O. J., Pantaleo, G., Lam, G. K., Fauci, A. S. (1997) Studies on
lymphoid tissue from HIV-infected individuals: implications for the design
of therapeutic strategies. Springer Semin. Immunopathol. 18, 305–322.
42. Schrager, L. K., Fauci, A. S. (1995) Human immunodeficiency virus.
Trapped but still dangerous. Nature 377, 680 681.
43. Germinario, R. J., Colby-Germinario, S. P. (2003) The effect of human
immunodeficiency virus-1 protease inhibitors on the toxicity of a variety of
cells. In Vitro Cell. Dev. Biol. Anim. 39, 275–279.
44. Fontana, G., Licciardi, M., Mansueto, S., Schillaci, D., Giammona, G.
(2001) Amoxicillin-loaded polyethylcyanoacrylate nanoparticles: influ-
ence of PEG coating on the particle size, drug release rate and phagocytic
uptake. Biomaterials 22, 2857–2865.
45. Cui, Z., Hsu, C. H., Mumper, R. J. (2003) Physical characterization and
macrophage cell uptake of mannan-coated nanoparticles. Drug Dev. Ind.
Pharm. 29, 689 –700.
10 Journal of Leukocyte Biology Volume 80, November 2006 http://www.jleukbio.org
... To address these problems, the same bone marrow and blood monocytes, tissue macrophages, microglia, and dendritic cells, which serve as targets, reservoirs, and vehicles for HIV dissemination, can be used as vehicles for antiretroviral therapy (ART). A nanoparticle indinavir (NP-IDV) formulation packaged into carrier bone marrow-derived macrophages (BMMs) was developed (Dou et al 2006). Drug distribution and disease outcomes were assessed in immune-competent and HIV-1-infected humanized immunedeficient mice, respectively. ...
... In another study where BMM were used as carriers for NP-IDV, the cell distribution was monitored by SPECT, T2 MRI, histology, and gamma-scintillation spectrometry (Gorantla et al 2006). BMM labeled with super paramagnetic iron oxide and/or 111 indium oxine were infused intravenously into naive mice. ...
Technical Report
Full-text available
This report reviews the current state-of-art of antiviral approaches including vaccines, pharmaceuticals and innovative technologies for delivery of therapeutics. The introduction starts with a practical classification of viral diseases according to their commercial importance. Various antiviral approaches are described including pharmaceuticals and molecular biological therapies such as gene therapy and RNA interference (RNAi) as well as vaccines for virus infections. Expert opinion is given about the current problems and needs in antiviral therapy. SWOT (strengths, weaknesses, opportunities and threats) analysis of antiviral approaches is presented against the background of concept of an ideal antiviral agent. A novel feature of this report is the use of nanotechnology in virology and its potential for antiviral therapeutics. Interaction of nanoparticles with viruses are described. NanoViricides are polymeric micelles, which act as nanomedicines to destroy viruses. Various methods for local as well as systemic delivery of antiviral agents and vaccines are described. Nanobiotechnology plays an important role in improving delivery of antivirals. Advantages and limitations of delivery of gene-based, antisense and RNAi antiviral therapeutics are discussed. Anti-influenza measures applicable to human as well as avian forms are described including the recent epidemic of swine flu. Resistance can develop against neuraminidase inhibitors although it is less than that with adamantanes. Considering these problems, there is need for a more effective agent. Investigations into alternative anti-influenza target will probably expand in the coming years. These include the development of mechanisms to inhibit fusion between the virus envelope and the cell membrane. After a discussion of current therapies of AIDS/HIV and their limitations, new strategies in development of antiviral agents are described. Drug resistance and toxicities are emerging as major treatment challenges. Based on a review of technologies and drugs in development, it can be stated that there are good prospects are of finding a cure for HIV/AIDS in the next decade. Hepatitis viruses are described with focus on hepatitis C virus (HCV) and hepatitis B virus (HBV). Despite the presence of numerous drug candidates in the anti-HCV pipeline, and the commitment of major R&D resources by many pharmaceutical companies, it might still take several years for any new anti-HCV drugs to reach the market. Although many companies are focusing their efforts on developing viral inhibitors, cellular targets in the host are beginning to emerge as attractive possibilities because they might enable the development of broad-spectrum antiviral drugs with less chance for developing viral resistance. Various commercially important viruses include herpes simplex (HSV) and human papilloma virus (HPV). There a number of treatments but HSV is not destroyed completely and remains dormant and activates from time to time to cause various clinical manifestations. There is discussion about the role of HPV in cervical cancer and vaccines available now seem to be adequate in preventing HSV-induced cervical cancer. There is no effective vaccine for respiratory syncytial virus (RSV) although monoclonal antibody (MAb) treatment is useful for prophylaxis and reducing the clinical manifestations. There is a need for an agent to eliminate this virus. Various viruses that either occur in epidemics or in tropics and some naturally emerging infectious diseases are described, e.g. viral hemorrhagic fevers such as dengue and West Nile virus infection. These are a constant threat and impossible to anticipate. Some of these lack antiviral agents or vaccines for prevention. Although these include some of the most serious viral disorders, the development of antiviral agents for these is not commercially attractive. Current research and approaches to these virus infections, particularly the current pandemic of COVID-19, are discussed. There are over 110 drugs and vaccine candidates in development of which 3 have been approved by the FDA and other health authorities around the world. Vaccination is being carried out in several countries Markets for antivirals are considered according to viruses and diseases caused by them and also according to management approaches: antiviral drugs, vaccines, MAbs and innovative approaches that include immunological and use of other technologies such as gene therapy, antisense, RNAi and nanobiotechnology. Antiviral markets are estimated starting with 2020 with projections up to the year 2030. Profiles of 197 companies that are involved in developing various technologies and products are profiled and with 182 collaborations. These include major pharmaceutical companies (12), Biopharmaceutical companies with antiviral products (87), Antiviral drug companies (26) as well as viral vaccine companies (71). The report is supplemented with 58 tables, 17 figures and 550 references from the literature.
... Mononuclear phagocyte ghost cells, including monocytes in the blood, macrophages in the tissues, microglial, and dendritic cells in bone marrow, are being employed as covert delivery vehicles for antiretroviral medicines to HIV latent forms. Magnetic resonance imaging (MRI) was used to examine the distribution of indinavirloaded bone marrow macrophages and SPIONs throughout the body [79]. ...
Article
Full-text available
Globally infectious diseases are the leading risk factor for death, with viruses having a particularly large influence on healthcare and socioeconomic development. Additionally, the fast development of medication resistance to presently available medicines, as well as harmful side effects from long-term usage is a major public health concern. Nanomedicine technology is developing as a novel technique capable of overcoming the limitations of molecular treatments and increasing their anti-infective activity by site-specific delivery to infection sites, lowering off-target effects and/or control of resistance emergence. NPs have distinctive physical features that are important in administration of drugs. Nanotechnologies have therapeutic applications and the potential to aid in the detection of viruses and viral diseases in the laboratory. Virus-like particles (VLPs) have minimal side effects and are safe and non-toxic, it has an interior hollow area; they are often used as nano-drug carriers. Up to date biopharmaceutical engineering is becoming a prominent topic.
... Shao et al. developed a self-guided biohybrid micromotor using natural neutrophils [55] that inherits the intrinsic chemotactic capacity of neutrophils and moves autonomously along a chemoattractant gradient for targeted drug transport. There are also differentiated cells like those derived from monocytes (part of the innate immune system): macrophages [83][84][85]. Munerati et al. reported for the first time the drug encapsulation capacity of macrophages [86], using their most typical characteristics of phagocytosis of microorganisms and other particles for the transport of antiretroviral drugs to active viral replication sites, which can play an important role in the treatment of cancer and HIV-associated neurocognitive disorders. In addition, Lee et al. developed a hybrid immune cell-based microrobot [87] using a click-response-assisted immune cell targeting strategy to deliver drug-laden nanoparticles deep into the avascular regions of tumors, yielding promising in vivo results, which are attractive for increasing penetration and accumulation into tumors/tissues. ...
Article
Full-text available
Single-cell microrobots are new microartificial devices that use a combination of single cells and artificial devices, with the advantages of small size, easy degradation and ease of manufacture. With externally driven strategies such as light fields, sound fields and magnetic fields, microrobots are able to carry out precise micromanipulations and movements in complex microenvironments. Therefore, single-cell microrobots have received more and more attention and have been greatly developed in recent years. In this paper, we review the main classifications, control methods and recent advances in the field of single-cell microrobot applications. First, different types of robots, such as cell-based microrobots, bacteria-based microrobots, algae-based microrobots, etc., and their design strategies and fabrication processes are discussed separately. Next, three types of external field-driven technologies, optical, acoustic and magnetic, are presented and operations realized in vivo and in vitro by applying these three technologies are described. Subsequently, the results achieved by these robots in the fields of precise delivery, minimally invasive therapy are analyzed. Finally, a short summary is given and current challenges and future work on microbial-based robotics are discussed.
... MONPs can be programmed to suppress or stimulate immunity. Iron oxide NPs served as vaccine transporters with established therapeutic effects, and they were also used to deliver antiretroviral medications to an inactive form of HIV (Gorantla et al., 2006). Citrate-coated MnFe 2 O 4 NPs can aid in the formation of a specific immune response (Neto et al., 2018). ...
... In addition to coating, nanocrystals can be internalized by cells for improved drug release and distribution in vivo [155][156][157][158]. Indinavir nanocrystals were packed into bone marrow-derived macrophages by phagocytosis [157]. ...
Article
Nanocrystals have contributed to exciting improvements in the delivery of poorly water-soluble drugs. The biological and intracellular fates of nanocrystals are currently under debate. Due to the remarkable commercial success in enhancing oral bioavailability, nanocrystals have originally been regarded as a simple formulation approach to enhance dissolution. However, the latest findings from novel bioimaging tools lead to an expanded view. Intact nanocrystals may offer long-term durability in the body and offer drug delivery capabilities like those of other nano-carriers. This review renews the understanding of the biological fates of nanocrystals administered via oral, intravenous, and parenteral (e.g., dermal, ocular, and pulmonary) routes. The intracellular pathways and dissolution kinetics of nanocrystals are explored. Additionally, the future trends for in vitro and in vivo quantification of nanocrystals, as well as factors impacting the biological and intracellular fates of nanocrystals are discussed. In conclusion, nanocrystals present a promising and underexplored therapeutic opportunity with immense potential.
... The presence of the PFC-labeled BMM in the liver, lungs, and spleen was readily apparent by 19 F-MRI and confirmed by ex-vivo 19 F-NMR spectroscopy. This pattern of accumulation is reminiscent of the one previously reported for labeled BMM tracked by singlephoton emission computed tomography (SPECT) and T 2 *-weighted MRI, where the cells were primarily located in the liver and spleen after initially being found mainly in the lung [25]. The similar pattern suggests that these two different labeling methods do not affect the BMM migration behavior, but a functional effect due to the labeling cannot be ruled out. ...
... Iron oxide NPs were used as undercover carriers to deliver anti-retroviral drugs to a latent form of HIV while reporting the localization of drugs owing to their contrasting properties [96]. A complex anti-HIV drug, enfuvirtide that is unable to cross the blood-brain barrier (BBB), was able to cross it when the drug was loaded on a PMA amphiphilic polymer-coated over iron oxide NPs [97]. ...
Article
Full-text available
The development of new nanomaterials with high biomedical performance and low toxicity is essential to obtain more efficient therapy and precise diagnostic tools and devices. Recently, scientists often face issues of balancing between positive therapeutic effects of metal oxide nanoparticles and their toxic side effects. In this review, considering metal oxide nanoparticles as important technological and biomedical materials, the authors provide a comprehensive review of researches on metal oxide nanoparticles, their nanoscale physicochemical properties, defining specific applications in the various fields of nanomedicine. Authors discuss the recent development of metal oxide nanoparticles that were employed as biomedical materials in tissue therapy, immunotherapy, diagnosis, dentistry, regenerative medicine, wound healing and biosensing platforms. Besides, their antimicrobial, antifungal, antiviral properties along with biotoxicology were debated in detail. The significant breakthroughs in the field of nanobiomedicine have emerged in areas and numbers predicting tremendous application potential and enormous market value for metal oxide nanoparticles.
Article
Full-text available
The concept of targeted drug delivery can be described in terms of the drug systems’ ability to mimic the biological objects’ property to localize to target cells or tissues. For example, drug delivery systems based on red blood cells or mimicking some of their useful features, such as long circulation in stealth mode, have been known for decades. On the contrary, therapeutic strategies based on macrophages have gained very limited attention until recently. Here, we review two biomimetic strategies associated with macrophages that can be used to develop new therapeutic modalities: first, the mimicry of certain types of macrophages (i.e., the use of macrophages, including tumor-associated or macrophage-derived particles as a carrier for the targeted delivery of therapeutic agents); second, the mimicry of ligands, naturally absorbed by macrophages (i.e., the use of therapeutic agents specifically targeted at macrophages). We discuss the potential applications of biomimetic systems involving macrophages for new advancements in the treatment of infections, inflammatory diseases, and cancer.
Chapter
NeuroAIDS is a neuropsychiatric disorder primarily caused by chronic human immunodeficiency virus (HIV). The most common neurological complications during NeuroAIDS are HAD (HIV-associated dementia) and HAND (HIV-associated neurocognitive disorder). HAART (highly active antiretroviral therapy) is a group of ARV (antiretroviral) drugs used to inhibit the key proteins involved in HIV replication. ARV drugs are extensively used to decrease the viral load and risk of opportunistic infections and to prolong survival of HIV-infected individuals. Significant advancement in antiretroviral (ARV) drugs has been achieved. However, the elimination of HIV from the CNS remains a difficult task. The complex structure of blood-brain barrier (BBB), high plasma protein binding, low patient compliance, and poor pharmacokinetics and biodistribution are the major limitations which restrict the effective concentration of drug to the CNS resulting in low efficacy of HAART in the CNS. Development of novel nanotechnology-based drug delivery methods for ARV drugs can increase the effectiveness of regimen with fewer side effects and better patient compliance which reduces financial load of healthcare system. The present article discusses about the nanotechnology-based approaches that can improve the delivery of ARV, called nano-ART. Nano-ART has excellent BBB permeability and improved pharmacokinetics and biodistribution that have significant clinical advantages to cure the CNS infection of HIV.
Article
Full-text available
In this paper, an overview of the pharmacokinetics of currently available antiretroviral drugs is provided. Included in this article are the agents zidovudine, stavudine, zalcitabine, lamivudine, didanosine, abacavir, nevirapine, delavirdine, efavirenz, saquinavir, indinavir, ritonavir and nelfinavir. Key pharmacokinetic parameters, drug penetration in body compartments and drug interactions are discussed for each agent.
Article
Full-text available
A method is described for the culture of mononuclear phagocytes in suspension by incubation on a Teflon film to which the cells do not adhere. The characteristics of peritoneal macrophages, bone marrow mononuclear phagocytes, macrophage cell lines, and fibroblasts cultured in this way are similar to those observed after culture on glass or plastic surfaces. Culture of mononuclear phagocytes in Teflon film dishes has three important advantages: the cells can be easily harvested without damage, recovery is almost complete, and the cells are not functionally impaired. Thus, this method makes it possible to use cultured mononuclear phagocytes for many studied that could previously only be done in freshly collected cells.
Book
Mononuclear phagocytes, which include macrophages, monocytes and their precursor cells, are the most important cells in the host defence against micro-organisms and tumor cells. During the last twenty-five years research on the biology of mononuclear phagocytes has increased tremendously. This motivated Professor R. van Furth to organize five international conferences on this subject in Leiden, the Netherlands. The edited proceedings of these meethings were published: in 1970 Mononuclear Phagocytes; in 1975 Mononuclear Phagocytes in Immunity, Infections and Pathology; in 1980 Mononuclear Phagocytes -- Functional Aspects; and in 1985 Mononuclear Phagocytes -- Characteristics, Physiology and Function. Reviews of these volumes, published in international journals, praised them as the most up-to-date state of the art publications. The publication of 1991 includes 88 chapters written by more than 200 authors.
Article
The results of a new test using indium oxine in the diagnosis of postoperative infection are reported. Indium-111 was used to label autologous polymorphonuclear leukocytes, which when reinjected migrate to sites of infection and inflammation. Standard scintigraphy localizes the labeled inflammatory cells at these sites. Sixty-six scans were performed in 43 surgical patients. Thirty-seven scans were categorized as true-positive; 19 scans were categorized as true-negative. Therefore, the accuracy rate was 85%. Two scans (3%) in one patient represented false-positive results. Two scans (3%) were positive for inflammation but there was no infection present; this group was denoted as equivocal. Six scans (9%) were false-negative; false-negative scans are more likely in old lesions with poor blood supply and in areas that overlap regions of normal uptake. The noninvasive nature of the test, high accuracy rate, and ease of administration make it a potentially useful tool in the diagnosis of postoperative infection.
Article
Determine the in vivo localization pattern of indium-111-labeled lymphocytes after a standardized extremity injury or standardized laparotomy and after sterile inflammation of the central nervous system. Prospective animal study with concurrent controls. Animal research laboratory. Male Lewis rats weighing 150-175 g. Indium-111-labeled splenic lymphocytes were injected into animals after a standardized hind limb trauma or laparotomy and after induction of sterile central nervous system inflammation. Lymphoid and non-lymphoid organs were removed at fixed intervals after lymphocyte injection and the proportion of injected lymphocytes/gram of tissue was determined using a quantitative radionuclide calculation. Results from treated animals were compared with results from untreated control animals. Muscle injury caused early localization of lymphocytes to injured hind limbs, liver, and spleen compared with controls, whereas laparotomy decreased lymphocyte localization in the thymus and colon. Encephalitis increased localization to the central nervous system with no effect on other tissues. These results identify a sensitive method to track in vivo leukocyte localization and specifically demonstrate that lymphocyte localization is altered in both traumatic and nontraumatic models of inflammation.
Article
Lymphoid tissue is a major reservoir of human immunodeficiency virus (HIV) infection in vivo. In addition, the lymphoid microenvironment provides a replicative advantage to the virus in that it provides a milieu of activated target cells that allows for efficient virus spread. The process of mobilization and activation of immune competent cells directed against the virus paradoxically contributes to the propagation of virus replication. Disruption of the lymphoid microenvironment during the progression of HIV disease is a poorly understood process, which may be of considerable importance pathogenically. Studies of lymph node biopsy samples taken 8 weeks apart from individuals who did not undergo any change in their therapeutic regimen (i.e., patients who either remained untreated or remained on their ongoing nucleoside analogue reverse transcriptase inhibitor monotherapy regimen) revealed little change in histopathology or viral load over the 8-week period. These results with successive lymph node biopsy samples taken from different sites indicate that an isolated lymph node biopsy accurately reflects the pathologic process associated with HIV infection and that this process diffusely involves the lymphoid system. Treatment with reverse transcriptase inhibitor monotherapy of patients in relatively early stage HIV disease had no detectable impact on the viral load in lymphoid tissue, suggesting the need to investigate more potent antiretroviral regimens during this stage of disease. Among patients with moderately advanced HIV disease, switching to combination therapy from a monotherapy regimen resulted in decreased viral replication in lymph nodes; this effect was associated with decreases in plasma viremia. Despite the fact that measures of viral replication decreased significantly, the net frequency of HIV-infected cells in peripheral blood and lymph nodes remained unchanged. Potent antiretroviral drug combinations may be capable of profound and long-term down-regulation of plasma viremia. It will be essential to monitor the status of viral trapping, viral burden, and viral replication within lymphoid tissue during treatment with such drugs to determine accurately their true potential for impact on these key features of HIV pathogenesis.
Article
Rationale and objectives: We examined the effects of arterial ischemia on the phagocytic activity of the hepatic macrophage-monocytic phagocytic system (MMPS). Methods: Six minipigs were studied before and 24 hr after complete arterial devascularization of the liver. Magnetic resonance (MR) imaging was performed at 1.5 T using superparamagnetic iron oxide (SPIO) particles (18 mumol Fe/kg body weight) as an MMPS-specific contrast agent. Hepatobiliary scintigraphy, measurements of serum liver enzymes, and histology also were obtained. Results: On MR imaging, the postcontrast-to-precontrast ratios of the arterially devascularized livers were significantly higher than the corresponding baseline values (p < .01). The greatest difference (52%) between the baseline and the postoperative values was observed on gradient-echo (GE) images. Scintigraphy, laboratory analyses, and histology results indicate that the MR imaging findings were probably predominantly attributable to a reduction in phagocytic activity of the hepatic MMPS. Conclusion: SPIO particles have already proved useful for improving detection of liver neoplasms on MR imaging, but they also may provide a novel way of evaluating the function of the hepatic MMPS in liver diseases.
Article
The purpose of this phase I study was to evaluate the toxicity and biological activity of autologous blood-derived macrophages activated ex-vivo with recombinant human interferon γ (rhuIFNγ) [monokine-activated killer (MAK) cells] and administered intravenously to 11 lung cancer patients once a week for 6 consecutive weeks. Peripheral blood monocytes were collected by leukapheresis and then purified by counterflow elutriation. The MAK cells were generated by culturing the purified monocytes in Teflon bags for 7 days and adding rhuIFNγ to the cultured cells for the last 18 h. These MAK cells expressed differentiation-associated surface antigen MAX1, and were cytotoxic in vitro against tumour cell line U937. The MAK cells were infused at dose levels from 1 × 107 to 5 × 108 on an intrapatient dose-escalating schedule. No severe adverse side-effects occurred. Toxicity was mild to moderate [primarly fever (75%) and chills (32%)], non-dose-dependent, and non-cumulative. No consistent change in haemostatic function, or liver or renal function was observed. Dose-limiting toxicity was not reached at 5 × 108 cells (optimal dose reproduced for each patient). The maximum tolerated dose was not determined. The immunomodulatory activity of i.v. infused MAK cells was demonstrated both in vivo by significant increases in granulocyte count and neopterin level in the patients' peripheral blood postinfusion and in vitro by secretory products (IL-1. TNFα, neopterin, and thromboplastin-like substance) in the culture supernatants. The in vivo traffic patterns of autologous MAK cells labelled ex-vivo with111In oxine were studied in 7 patients. Gamma imaging showed an immediate but transient lung uptake (