You are on page 1of 10

HUMAN GENE THERAPY 12:1551–1558 (August 10, 2001)

Mary Ann Liebert, Inc.

Sustainable Systemic Delivery via a Single Injection of


Lentivirus into Human Skin Tissue

SEUNG-CHEOL BAEK,1 QUN LIN, PAUL B. ROBBINS, HONGRAN FAN, and PAUL A. KHAVARI

ABSTRACT

The skin offers a tissue site accessible for delivery of gene-based therapeutics. To develop the capability for
sustained systemic polypeptide delivery via cutaneous gene transfer, we generated and injected pseudotyped
HIV-1 lentiviral vectors intradermally at a range of doses into human skin grafted on immune-deficient mice.
Unlike Moloney murine leukemia virus (MLV)-based retrovectors, which failed to achieve detectable cuta-
neous gene transfer by this approach, lentivectors effectively targeted all major cell types within human skin
tissue, including fibroblasts, endothelial cells, keratinocytes, and macrophages. After a single injection, lentivec-
tors encoding human erythropoietin (EPO) produced dose-dependent increases in serum human EPO levels
and hematocrit that increased rapidly within one month and remained stable subsequently. Delivered gene
expression was confined locally at the injection site. Excision of engineered skin led to rapid and complete
loss of human EPO in the bloodstream, confirming that systemic EPO delivery was entirely due to lentiviral
targeting of cells within skin rather than via spread of the injected vector to visceral tissues. These findings
indicate that the skin can sustain dosed systemic delivery of therapeutic polypeptides via direct lentivector in-
jection and thus provide an accessible and reversible approach for gene-based delivery to the bloodstream.

OVERVIEW SUMMARY sues, the skin is readily available for direct topical and injectable
gene transfer in a minimally invasive and uncomplicated fash-
Direct vector administration to skin avoids the costly, ion. Direct gene delivery approaches to skin include topical ap-
painful, and scarring process of skin harvesting, in vitro plication (Li and Hoffman, 1995; Fan and Khavari, 1999; Do-
gene transfer, and regrafting. To develop an approach to mashenko et al., 2000), intradermal injection (Hengge et al.,
durable direct cutaneous gene transfer, we injected lentivec- 1995; Choate and Khavari, 1997), bioplastic particle accelera-
tors into human skin tissue on immune-deficient mice at a tion (Wang et al., 1999; Davidson et al., 2000), and vector in-
range of doses. Lentivectors successfully targeted all major fusion onto wounded skin ( Lu et al., 1997; Ghazizadeh et al.,
cell types within the epidermis and dermis, supporting vec- 1999). In addition to ease of administration, the skin is easily
tor dose-dependent and stable delivery of erythropoietin to subjected to direct clinical inspection following gene transfer
the bloodstream. Gene delivery remained localized in the with uncomplicated removal of genetically engineered tissue
skin and could be readily removed by punch biopsy of the achieved by simple excision if needed. These tissue character-
injected site. These data establish an approach to durable istics have facilitated progress in gene transfer for primary skin
systemic polypeptide delivery via a single injection of disorders, including the recessive ichthyoses (Choate et al.,
lentivector into human skin tissue. 1996a,b; Choate and Khavari, 1997; Freiberg et al., 1997a) and
epidermolysis bullosa subtypes (Seitz et al., 1999; Dellambra
et al., 2000; Robbins et al., 2001).
INTRODUCTION In addition to primary skin diseases, cutaneous gene trans-
fer offers promise for the treatment of systemic disorders re-

A S THE MOST ACCESSIBLE OF BODY ORGANS , the skin provides


an attractive tissue for therapeutic gene transfer (Cao et
al., 2000; Ghazizadeh and Taichman, 2000; Khavari, 2000;
sponding to delivery of polypeptides to the circulation because
the skin is richly vascularized. A range of skin-produced pro-
teins have been demonstrated to reach the bloodstream follow-
Uitto and Pulkkinen, 2000; Vogel, 2000). Unlike visceral tis- ing a variety of approaches to gene insertion into cells of both

Veterans Affairs Palo Alto Healthcare System and Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305.
1Present address: Department of Dermatology, Catholic University College of Medicine, Seoul, Korea.

1551
1552 BAEK ET AL.

the epidermis and the dermis or via addition of unmodified hu- hematocrit in a vector dose-dependent manner. These findings
man skin elements. Such polypeptides include growth hormone identify a promising approach to minimally invasive direct gene
(Morgan et al., 1987; Teumer et al., 1990; Jensen et al., 1994; transfer to skin for the treatment of systemic disease.
Wang et al., 1997), apolipoprotein E (Fenjves et al., 1989), Fac-
tor IX (Gerrard et al., 1993), interleukin-10 (Meng et al., 1998),
and transferrin (Petersen et al., 1995). While convincingly
MATERIALS AND METHODS
demonstrating the potential for cutaneous gene transfer to
achieve systemic delivery, these studies have predominantly re-
Vectors
lied on ex vivo gene delivery to skin. Ex vivo gene transfer to
skin requires biopsy for tissue harvesting, then growth and gene The self-inactivating lentiviral transfer vector backbones in
transfer in vitro followed by the painful and generally scarring which an internal cytomegalovirus (CMV) promoter drives ex-
process of regrafting and is thus a costly, complicated and trau- pression of either lacZ (Dull et al., 1998) or green fluorescent pro-
matic process. Widespread use of the skin for genetic treatment tein (GFP) (Naldini et al., 1996) have been described. The full-
of systemic disease will therefore require improved approaches. length human EPO cDNA (gift of P. Aebischer) was digested with
To make skin gene transfer widely applicable to a range of BamHI and XhoI and subcloned into the BamHI and XhoI sites
human systemic diseases, new capabilities for minimally inva- of PHR-CMV-GFP (Naldini et al., 1996) to generate PHR-CMV-
sive direct cutaneous gene delivery are needed. These involve EPO which contains 350 bp of gag as well as env sequences en-
sustainability, efficiency, and control of gene transfer to skin. compassing the Rev-responsive element (RRE). Lentivirus was
To date, no approach or vector yet studied has fully met these produced by transient co-transfection of human 293T cells with
criteria after direct administration to skin. Plasmid DNA, per- three plasmids. A total of 15 mg of transfer vector, 10 mg of the
haps the most well studied, displays very low efficiencies of pCMVDR8.2 packaging plasmid, and 5 mg of pCI-VSV-G, which
gene transfer when topically applied or injected and is subse- encodes the vesicular stomatitis virus envelope G (VSV-G) gly-
quently lost within days due to lack of integration (Hengge et coprotein, were transfected into 5 3 106 293T cells grown in
al., 1996; Choate and Khavari, 1997; Fan and Khavari, 1999). DMEM with 10% fetal bovine serum (FBS) in 10-cm dishes us-
While vastly more efficient, adenoviral gene transfer also suf- ing FuGene 6 (Roche, Indianapolis, IN) according to the manu-
fers from a lack of durability due to a failure to integrate into facturer’s instructions. Conditioned medium was replaced the day
target cells and immunogenicity ( Lu et al., 1997; Ghazizadeh following transfection and supernatant harvested 48 hr post-trans-
et al., 1999). Moloney murine leukemia virus (MLV) retro- fection. Supernatant was passed through a 0.45-mm filter then con-
viruses are also limited in that detectable levels of direct skin centrated by ultracentrifugation at 40,000 3 g for 2 hr. Pellets
gene transfer have only been achieved when the virus is in- were resuspended in media and then concentrated by precipita-
jected beneath hemorrhagic eschar after the skin has been sub- tion in a final concentration of 10% polyethylene glycol and pel-
jected to a tissue destructive wounding procedure to stimulate leted by centrifugation for 20 minutes at 5000 3 g. The final vi-
cellular proliferation (Ghazizadeh et al., 1999). Adeno-associ- ral pellet was resuspended for 4 hr at 4°C in phosphate-buffered
ated virus (AAV) vectors are capable of infecting cells of saline (PBS) to yield concentrations of 10- to 200-fold. Viral vec-
murine skin and achieving systemic protein delivery. However, tor stocks were titered by infecting target cells with serial dilu-
this is likely to be of limited clinical relevance because AAV tions in media supplemented with Polybrene at a concentration of
appears capable of sustaining delivery only via retention in the 8 mg/ml. Expression of b-galactosidase was analyzed by staining
mouse panniculus carnosus (Donahue et al., 1999), a muscular with X-gal (Sigma, St. Louis, MO.). Viral p24 antigen concen-
structure that does not exist in human skin. Thus, prior ap- trations were determined by HIV-1 core profile enzyme-linked
proaches to direct cutaneous gene transfer have one or more immunosorbent assay (ELISA; NEN Life Science Products,
shortcomings in meeting criteria for practically effective direct Boston, MA). In our studies, 1 mg of p24 equivalent corresponded
gene transfer to the skin. to approximately 2.5 3 106 infectious viral particles.
Among the vector platforms available, HIV-1 lentiviral vec-
tors are attractive candidates because of their ability to initiate Cells and gene transfer
and sustain gene delivery efficiently in both dividing and non-
Primary human skin fibroblasts were isolated and maintained
dividing cells after injection into neural and muscle tissue (Nal-
in Dulbecco’s modified Eagle medium (DMEM) with 10% FBS
dini et al., 1996; Zufferey et al., 1998; Kafri et al., 1999; Lever
as described (Balcar et al., 1994; Freiberg et al., 1997b). Cells
et al., 1999). Among the systemic disorders potentially
were transduced in vitro in media supplemented with Polybrene
amenable to cutaneous gene transfer are those characterized by
at 8 mg/ml by addition of vector followed immediately by 90
inadequate blood levels of specific polypeptides, such as is seen
min of centrifugation at 1000 3 g at 32°C. Culture media of
in erythropoietin (EPO)-responsive anemia. Here we have de-
EPO transduced cells was changed 2 hr before collection of su-
livered HIV-1-based lentivectors encoding human EPO along
pernatants 48 hr following transduction.
with marker gene controls by a single intracutaneous injection
into full-thickness human skin tissue grafts on immune-defi-
Expression analysis
cient mice. We have used human skin as a target tissue to en-
hance the practical value of this approach because it differs in Human EPO was assayed by human species-specific ELISA
significant ways from mouse and other animal models, includ- using the human EPO Quantikine IVD Kit (R&D Systems, Min-
ing at the levels of anatomy and viral tropism. This gene de- neapolis, MN) according to the manufacturer’s recommenda-
livery approach targeted all of the major cell types within skin tions. Blood obtained at various times before and after vector
with achievement of sustained EPO delivery and increased administration was analyzed for hematocrit using an automated
SYSTEMIC EPO DELIVERY VIA HUMAN SKIN 1553

hematology analyzer (Cell-Cyn 3500, Abbott Laboratories, Ab- 1997), however, failed to produce detectable immunohisto-
bott Park, IL). For immunohistochemistry, 5-mm skin cryosec- chemical evidence of gene transfer when equivalent viral doses
tions were incubated with human species-specific antibodies at were injected (data not shown). Lentivirus-mediated gene ex-
1:50 dilution in PBS. Antibodies used recognized cell-surface pression persisted for at least 6 months (the duration of analy-
proteins expressed on human skin macrophages (F4/80, Serotec, sis) and was focally confined within the skin around the nee-
Oxford, UK), endothelial cells (Factor VIII, Dako, Carpenteria, dle tract site of injection. To exclude the possibility that this
CA), and fibroblasts (a5 integrin, gift of J. Lie, Stanford, CA). could be due to injection of protein present in the viral super-
Tetramethylrhodamine isothiocyanate (TRITC)-labeled sec- natant, supernatants generated without Gag and Pol proteins
ondary antibodies (Sigma, St. Louis, MO) were used at 1:500 were also injected. These failed to show any evidence of marker
dilution in PBS. Co-staining was performed using fluorescein gene expression (data not shown), confirming that observed ex-
isothiocyantate (FITC)-labeled antibodies to b-galactosidase at pression was due to viral infection of cells within skin.
a 1:50 dilution (Rockland, Gilbertsville, PA). Sections were In epidermal keratinocytes, marker gene expression was of-
then mounted and visualized using dual-color fluorescence mi- ten evident in thin columns (Fig. 1), consistent with targeting
croscopy. For histochemical detection of lacZ gene expression, of long-lived progenitor cells at the basement membrane zone
grafted skin was biopsied and then frozen immediately on dry with subsequent expression in their outwardly migrating prog-
ice. The 5-mm cryosections were fixed in 0.25% glutaraldehyde eny. lacZ expression was also detected in major cell types
and stained with X-Gal as described (Deng et al., 1998). within the dermis, including endothelial cells, fibroblasts, and
macrophages using human species-specific antibodies to mark-
Animal studies ers for these cell types (data not shown). Among transduced
cells, fibroblasts comprised approximately 50%, endothelial
Adult CB.17 scid/scid mice were anesthetized with tribro-
cells 25%, and macrophages 10%; the remainder of lacZ[1]
moethanol by intraperitoneal (i.p.) injection. 2.0 3 2.0-cm
cells did not appear to react with the cell-type-specific anti-
squares of full-thickness human skin were trimmed of subcuta-
bodies used and thus may represent other dermal cell types.
neous fat and transplanted onto the backs of recipient mice at a
site in which overlying murine skin had been excised down to
fascia as described (Choate et al., 1996b). Nonhealing grafts, Sustained systemic delivery of human erythropoeitin
which represented approximately 5% of the total, were excluded
After confirming the ability of HIV-1 lentivectors to effect
from subsequent use. Lentiviral vector in a total volume of 50
durable gene expression in significant numbers of cells within
ml was injected intradermally (i.d.) using a 30-gauge needle at
human skin tissue, we wished to test the capacity of this ap-
a single site in the center of the graft. Three months after graft-
proach to deliver therapeutic gene products to the bloodstream
ing, at a time when gene expression and cell division have re-
sustainably. To do this, we constructed and produced a VSV-G
covered to normal levels seen in undisturbed normal human skin
pseudotyped HIV-1-based vector encoding human EPO, a
(Medalie et al., 1996), mice were injected with either 1, 5 or 10
polypeptide widely used in the treatment of anemia. First, we
mg of p24 equivalent of lentivector encoding EPO (n 5 5 per
confirmed the ability of this lentivector to produce high levels
dose). Two mice were injected with lacZ control lentivector at
of secreted EPO in target cells from human skin grown in vitro.
the 10 mg of p24 equivalent dose. Adult CB.17 scid/scid mice
We infected primary skin fibroblasts and measured EPO se-
were also used for injection studies using wholly murine skin.
creted in the media 48 hr later. Gene transfer at multiplicities
of infection (MOI) of 15 was performed to achieve .99% ef-
ficiency lacZ marker gene transfer. In this setting, as few as
RESULTS
1 3 105 cells could produce EPO in a vector dose-dependent
manner at rates as high as 8.5 IU/hr (Fig. 2). This confirmed
Lentiviral gene transfer to human skin
the ability of lentivector to support EPO secretion in the pre-
To study the ability of lentiviral vectors to achieve cutaneous dominant cellular target type within skin.
gene transfer, we produced and injected VSV-G pseudotyped After confirming the effectiveness of this lentivector in pri-
HIV-1 lentiviral virus into full-thickness human skin grafted on mary cells from human skin in vitro, we wished to test its abil-
immune deficient CB.17 scid/scid mice. A self-inactivating ity to achieve sustained systemic delivery via cutaneous gene
HIV-1 transfer vector (Naldini et al., 1996; Dull et al., 1998;) transfer. We injected 1, 5, and 10 mg of p24 equivalent intra-
was used; it contained a 400-bp deletion in the U3 long termi- dermally into human skin grafted on severe immunodeficient
nal repeat (LTR), with transcription driven by internal CMV (SCID) mice. This produced a prolonged delivery of human EPO
promoter. This backbone transfer vector was used to deliver in the serum with a corresponding dose-dependentboost in hema-
lacZ and GFP marker genes directly to skin by injection into tocrit (Fig. 3 and Table 1). The well-described mortality over
the dermis, with five human tissue grafts injected with a 30- time characteristic of this grafting model was observed (Robbins
gauge needle for each vector. Marker gene expression was ob- et al., 2001) and accounted for the animals noted as “not done”
served in all cases for both GFP and lacZ vectors after injec- at each time point; however, certain trends were clear in the data.
tion of 5 mg of p24 viral equivalent, with expression evident in Although variability was seen between individual mice (Table
cells of both the dermis and epidermis (Fig. 1). Both lentivec- 1), the average magnitude of this response corresponded to the
tors produced equivalent levels of gene transfer when injected dose of vector initially administered. EPO delivery was stable for
into human skin at comparable doses. The IN-GFP MLV retro- the 6-month duration of the experiment, a time limit determined
vector that we have previously shown can drive robust gene ex- primarily by the viability of both test and control grafted im-
pression in skin tissue after ex vivo gene transfer (Deng et al., mune-deficient mice in our hands. In the single mouse that sur-
1554 BAEK ET AL.

FIG. 1. Lentiviral vector gene delivery to human skin tissue. A total of 5 mg of p24 equivalent of lentiviral vector encoding
lacZ was injected into full-thickness human skin grafted on SCID mice. Localization of transduced cells by X-Gal staining. Note
that the first low-power panel demonstrates expression tightly localized to the needle tract, whereas the second demonstrates a
more loosely grouped distribution of expressing cells within the dermis. The third higher-power panel displays lacZ 1 cells ly-
ing among dermis, whereas the fourth panel shows a column of engineered keratinocytes in the overlying epidermis (arrows).

vived into the eleventh month post-injection (mouse 10-2, Table well-vascularized dermis demonstrated lacZ expression local-
1), both EPO levels and hematocrit were stable at 25 mIU/ml and ized at the site of intradermal injection and not elsewhere in
72%, respectively. To quantitate the numbers of cells in skin skin away from the injection site or in visceral tissues (Fig. 1
transduced at the doses of lentivector producing these sustained and data not shown). Consistent with a lack of lentiviral dis-
EPO levels, we performed immunohistochemical analysis of se- semination, local punch biopsy excision of dermis and epider-
rial 5-mm tissue sections spanning the entire region of injected mis at the HIV-1 EPO skin injection site approximately 1 month
human skin tissue in multiple independently injected human skin after injection rapidly led to complete loss of detectable human
grafts. By this approach, we estimated that 3,343 6 1438 cells EPO in the serum (Fig. 4). The fact that nonexcised skin tissue
in human skin are transduced per mg of p24 equivalent of could support stable EPO expression for as long as 10 months
lentivector. This indicates that a relatively small number of sta- more, in contrast to this drop to undetectable levels following
bly engineered cells in skin can durably deliver systemically ac- excision, confirms the constrained localization of lentiviral gene
tive levels of proteins to the bloodstream. transfer to the skin and indicates that the observed targeted skin
cells are responsible for EPO reaching the bloodstream.
Confinement of gene transfer to skin and reversibility
An ideal feature of cutaneous gene transfer would be reten-
tion of genetically engineered cells in a localized area of skin. DISCUSSION
This would permit local control of gene delivery topically as
well as allow removal of all engineered cells in the event of a We have demonstrated gene delivery with durable systemic
hypersensitivity reaction to the delivered gene product. Did the expression via a single lentivector injection into human skin tis-
observed EPO expression meet this criteria? Intracutaneous in- sue. Advantages of this skin-targeted approach include the fact
jection of 1–10 mg of p24 equivalent HIV-1 lacZ targeting the that vector administration is minimally invasive and requires

A B

FIG. 2. Infection with a lentiviral vector encoding human EPO leads to dose-dependent EPO secretion in vitro. (A) Proviral
genome diagram of the PHR-CMV-EPO transfer vector. LTR, HIV-1 long terminal repeat; SD, splice donor; SA, splice accep-
tor; Ga, gag sequences; C, extended packaging sequences; CMV,799-bp cytomegalovirus immediate early gene promoter.
(B) Increasing amounts of EPO-encoding lentivector noted were used to infect primary human dermal fibroblasts in vitro. At
48 hr after infection, the presence and concentration of human EPO secreted into the media per hour was analyzed.
SYSTEMIC EPO DELIVERY VIA HUMAN SKIN 1555

A B

FIG. 3. Dose-dependent systemic delivery of human EPO and increase in hematocrit following a single intradermal lentivec-
tor injection in human skin tissue. Increasing amounts of EPO encoding lentivector were injected intradermally into full-thick-
ness human skin grafted onto SCID mice at time zero (n 5 5 independently grafted mice for each dose); lacZ lentivector at the
highest dose served as a control (n 5 2). (A) Levels of human EPO in serum as detected by human species-specific EPO ELISA.
(B) Hematocrit over time following injection.

only a single superficial cutaneous injection with a narrow- advantage of this approach is its durability. Although human
gauge needle. In addition, the localized nature of gene delivery skin/SCID mouse viability limited our studies to a maximum
was underscored by the fact that systemic gene product deliv- of generally 6 months, but as long as 11 months in a single sur-
ery could be completely terminated by removal of injected tis- viving mouse, it is possible that cutaneous gene transfer of an
sue. This ability to achieve rapid and total cessation of gene de- endogenous nonimmunogenic EPO gene to humans could re-
livery by skin biopsy excision of engineered cells offers sult in much longer gene expression. This possibility is sup-
advantages over gene transfer to visceral organs where com- ported by the fact that we observed no evidence of local tissue
plete removal of engineered cells either requires an invasive toxicity in skin after long-term lentiviral gene delivery as well
procedure or, in the case of hematopoietic lineages dissemi- as by the fact that gene expression generally reached a plateau
nated in the circulation, may be nearly impossible. A further within 1 month of injection that remained stable subsequently.

TABLE 1. EPO AND HEMATOCRIT V ALUES FOR INDIVIDUAL INJECTED MICE AT SPECIFIC TIME POINTS

Serum hEPO (mIU/ml) Hematocrit (%)

Dose Mouse Pre 2 wks 2 mo. 4 mo. 6 mo. Pre 2 wks 2 mo. 4 mo. 6 mo.
1 mg 1—1 ,2.5 ,2.5 ,2.5 ,2.5 ,2.5 42 44 44 41 40
1—2 ,2.5 ,2.5 ,2.5 ,2.5 ,2.5 44 47 43 44 43
1—3 ,2.5 3.3 2.8 ,2.5 ,2.5 41 50 49 47 49
1—4 ,2.5 4.9 3.7 2.8 3.4 42 53 51 48 48
1—5 ,2.5 5.3 6 4.2 5.5 45 54 55 51 50

5 mg 5—1 ,2.5 5 3.5 2.5 3.2 42 53 49 49 51


5—2 ,2.5 5.9 4.2 3.5 3.5 46 54 51 52 48
5—3 ,2.5 8.5 7.8 ND ND 44 55 56 ND ND
5—4 ,2.5 14.5 17.5 18.4 15.8 41 56 57 59 55
5—5 ,2.5 16.5 16 16 15 40 55 56 55 54

10 mg 10—1 ,2.5 17 16.5 ND ND 43 53 55 ND ND


10—2 ,2.5 24.3 30.5 25.5 26 42 58 64 70 68
10—3 ,2.5 25.5 28.5 25.5 27.7 45 57 60 70 65
10—4 ,2.5 34 40.4 36.8 39.5 44 60 72 76 72
10—5 ,2.5 49 55.8 ND ND 45 63 80 ND ND

LacZ C—1 ,2.5 ,2.5 ,2.5 ,2.5 ,2.5 42 40 42 42 40


C—2 ,2.5 ,2.5 ,2.5 ,2.5 ,2.5 43 39 40 39 41

These data represent the numbers used to generate the graphs in Fig. 3.
Serum hEPO and hematocrit levels in individual mice grafted with full-thickness human skin which received a single injec-
tion with the indicated HIV1 p24 equivalent doses of hEPO vector. These data are shown in graphical form in Fig. 3. ,2.5, Not
detected; ND, not done, due to the SCID mouse death commonly observed due to infection over time (Robbins et al).
1556 BAEK ET AL.

epidermis and dermis, we failed to observe any gene expression


after direct injection of AAV vector encoding lacZ into full-thick-
ness human skin tissue (data not shown). Thus assessment of in-
tracutaneous systemic gene delivery using AAV and other vec-
tors via human skin tissue awaits further development.
Dosing of serum EPO was achieved in a stable fashion that
correlated with the amount of vector administered. Such a sta-
ble and sustained level of gene expression may be useful in cer-
tain settings that do not require tight regulation, such as in de-
livery of Factor IX in hemophilia where there is a large
therapeutic window (Kay et al., 2000). Further refinements in
the ability to dose gene production via direct cutaneous trans-
fer are needed, however, for several reasons. First, there was
variability observed in delivered EPO, even among individual
mice receiving the same vector dose. Second, a number of other
applications will require the capacity for temporally regulated
dosing, such as for delivery of genes whose expression levels
FIG. 4. Loss of serum human EPO after excision of injected must be more precisely regulated in time. Among the most
skin. Four weeks following a single EPO lentivector injection, prominent of these is insulin for diabetes mellitus, where ex-
epidermis and dermis were removed by simple punch biopsy cessive amounts of delivered insulin could prove lethal in the
excision of the engineered central portion of grafted skin and form of hypoglycemic shock. A number of other applications,
human serum EPO levels were determined 10 days later.
however, including EPO-responsive anemias, could also benefit
from the capability for exogenous regulation. In the case of the
latter, we have observed that excessive delivery via skin clearly
Significantly, we failed to observe even short-term detectable leads to dramatically elevated hematocrits that may pose a ma-
lentiviral gene transfer in wholly murine skin using immune- jor risk of thrombosis. Of the 3 mice that died by 4 months of
deficient SCID animals for reasons that are unclear, pointing this study, one of these displayed a hematocrit of 80% and may
out an important difference between this commonly used model have died of this complication, although we could not confirm
animal and the goal human tissue target. Thus, the potential this on autopsy. Thus, given the fact that stable systemic de-
clinical utility of direct intracutaneous lentiviral injection for livery is possible via intracutaneous lentiviral injection, a ma-
systemic polypeptide delivery may be greater because we have jor next step would be the capacity for regulated production. In
used human target tissue, verifying transfer to human cells us- the case of the skin, such regulation may be approachable in a
ing species-specific antibodies. convenient fashion by topical regulation via application of com-
Systemically delivered gene products with significant bio- mercially available gene regulatory ligands, such as glucocor-
medical impact have been delivered by a number of approaches ticoids, retinoids, and vitamin D analogs.
in visceral tissues and skin. EPO is one of the best-studied pro- We have observed that pseudotyped HIV-1 lentivectors suc-
totype genes, and a variety of routes and vectors have been used cessfully target all of the major cell types within human skin
to achieve clinically significant delivery to the bloodstream. In tissue. Although the VSV-G envelope may not prove an ideal
the case of ex vivo gene delivery, the primary cell types used choice for direct human administration (DePolo et al., 2000),
have been myoblasts engineered with plasmid (Dalle et al., other envelopes with wide cellular tropism are available, in-
1999) and retroviral vectors (Hamamori et al., 1995; Bohl et al., cluding alphaviruses (Sharkey et al., 2001), gibbon ape leuke-
1997) as well as fibroblasts engineered with both (Serguera et mia virus (Stitz et al., 2000), and others. Which of the cell types
al., 1999). In the case of direct in vivo gene delivery, the pri- targeted contributed to EPO delivery to the bloodstream? While
mary tissue target has been the muscle, with plasmid (Abruzzese skin fibroblasts have been clearly shown capable of systemic
et al., 2000; Lemieux et al., 2000), AAV (Yan et al., 2000), and delivery by ex vivo studies in which pure populations were en-
adenovirus (Svensson et al., 1997) delivered by direct injection. gineered then regrafted (Petersen et al., 1995), it is also of in-
The mouse has been the primary animal model used with sev- terest that endothelial cells comprised approximately one-fourth
eral studies in rats, cats and primates (Svensson et al., 1997; of the cells targeted. This raises the possibility that endothe-
Beall et al., 2000; Rudich et al., 2000). In several cases, the fo- lium may represent a key site of production-delivery by direct
cus has been on regulated EPO delivery using either tetracy- secretion into the blood. Assessing the relative contributions of
clines (Bohl et al., 1997), progesterone analogs (Abruzzese et each cell type in human tissue poses formidable technical chal-
al., 2000) or rapamycin (Ye et al., 1999) for pharmacologic con- lenges because absolutely cell type specific targeting vectors
trol of regulated promoters. In the single study in which EPO-en- for all of these cells in skin have yet to be developed.
coding vectors were delivered directly to skin, an AAV vector These data indicate lentiviral vectors are capable of skin-lo-
was delivered to entirely murine tissue. However, gene expres- calized gene transfer achieving systemic delivery of therapeu-
sion in this work could not be detected in cells of dermis or epi- tic polypeptides after a single intradermal injection. The local-
dermis, and only in the mouse panniculus carnosus (Donahue et ized nature of this gene transfer approach opens the way for
al., 1999), a muscular structure deep in the tissue that does not future development of direct lentiviral gene transfer for primary
exist in human skin. Consistent with this lack of expression in the skin disorders, as well as additional systemic diseases.
SYSTEMIC EPO DELIVERY VIA HUMAN SKIN 1557

ACKNOWLEDGMENTS DONAHUE, B.A., MCARTHUR, J.G., SPRATT, S.K., BOHL, D., LA-
GARDE, C., SANCHEZ, L., KASPAR, B.A., SLOAN, B.A., LEE,
This work was supported by the USVA Office of Research Y.L., DANOS, O., and SNYDER, R.O. (1999). Selective uptake and
sustained expression of AAV vectors following subcutaneous deliv-
and Development and by National Institutes of Health grants
ery. J. Gene Med. 1, 31–42.
AR44012, AR45192. and AR43799 to P.A.K. We thank L. Nal-
DULL, T., ZUFFEREY, R., KELLY, M., MANDEL, R.J., NGUYEN,
dini for the HIV-1 transfer vector backbones, P. Aebischer for M., TRONO, D., and NALDINI, L. (1998). A third-generation
the human EPO cDNA, D. Kohn for helpful advice, and P. lentivirus vector with a conditional packaging system. J. Virol. 72,
Bernstein and N. Griffiths for expert administrative support. 8463–8471.
FAN, H., and KHAVARI, P.A. (1999). Genetic immunization via hair
follicles after topical application of naked DNA to normal skin. Na-
ture Biotech. 17, 870–872.
REFERENCES FENJVES, E.S., GORDON, D.A., PERSHING, L.K., WILLIAMS,
D.L., and TAICHMAN, L.B. (1989). Systemic distribution of
ABRUZZESE, R.V., GODIN, D., MEHTA, V., PERRARD, J.L., apolipoprotein E secreted by grafts of epidermal keratinocytes: im-
FRENCH, M., NELSON, W., HOWELL, G., COLEMAN, M., plications for epidermal function and gene therapy. Proc. Natl. Acad.
O’MALLEY, B.W., and NORDSTROM, J.L. (2000). Ligand-de- Sci. USA 86, 8803–8807.
pendent regulation of vascular endothelial growth factor and ery- FREIBERG, R.A., CHOATE, K.A., DENG, H., ALPERIN, E.S.,
thropoietin expression by a plasmid-based autoinducible GeneSwitch SHAPIRO, L.J., and KHAVARI, P.A. (1997a). A model of correc-
system. Mol. Ther. 2, 276–287. tive gene transfer in X-linked ichthyosis. Hum Molec Genet. 6, 937–
BALCAR, V.J., SHEN, J., BAO, S., and KING, N.J. (1994). Na(+)- 933.
dependent high affinity uptake of L-glutamate in primary cultures of FREIBERG, R.A., SPENCER, D.M., CHOATE, K.A., DUH, H.J.,
human fibroblasts isolated from three different types of tissue. FEBS SCHREIBER, S.L., CRABTREE, G.R., and KHAVARI, P.A.
Lett. 339, 50–54. (1997b). Fas signal transduction triggers either proliferation or apop-
BEALL, C.J., PHIPPS, A.J., MATHES, L.E., STROMBERG, P., and tosis in human fibroblasts. J. Invest. Dermatol. 108, 215–219.
JOHNSON, P.R. (2000). Transfer of the feline erythropoietin gene GERRARD, A.J., HUDSON, D.L., BROWNLEE, G.G., and WATT,
to cats using a recombinant adeno-associated virus vector. Gene Ther. F.M. (1993). Towards gene therapy for haemophilia B using primary
7, 534–539. human keratinocytes. Nature Genet. 3, 180–183.
BOHL, D., NAFFAKH, N., and HEARD, J.M. (1997). Long-term con- GHAZIZADEH, S., and TAICHMAN, L.B. (2000). Virus-mediated
trol of erythropoietin secretion by doxycycline in mice transplanted gene transfer for cutaneous gene therapy. Hum. Gene Ther. 11, 2247–
with engineered primary myoblasts. Nat Med. 3, 299–305. 2251.
CAO, T., WANG, X.J., and ROOP, D.R. (2000). Regulated cutaneous GHAZIZADEH, S., HARRINGTON, R., and TAICHMAN, L. (1999).
gene delivery: the skin as a bioreactor. Hum. Gene Ther. 11, 2297– In vivo transduction of mouse epidermis with recombinant retrovi-
2300. ral vectors: implications for cutaneous gene therapy. Gene Ther. 6,
CHOATE, K.A., and KHAVARI, P.A. (1997). Direct cutaneous gene 1267–1275.
delivery in a human genetic skin disease. Hum. Gene Ther. 8, 1671– HAMAMORI, Y., SAMAL, B., TIAN, J., and KEDES, L. (1995). Myo-
1677. blast transfer of human erythropoietin gene in a mouse model of re-
CHOATE, K.A., KINSELLA, T.M., WILLIAMS, M.L., NOLAN, G.P., nal failure. J. Clin. Invest. 95, 1808–1813.
and KHAVARI, P.A. (1996a). Transglutaminase 1 delivery to lamel- HENGGE, U.R., CHAN, E.F., FOSTER, R.A., WALKER, P.S., and
lar ichthyosis keratinocytes. Hum. Gene Ther. 7, 2247–2253. VOGEL, J.C. (1995). Cytokine gene expression in epidermis with
CHOATE, K.A., MEDALIE, D.A., MORGAN, J.R., and KHAVARI, biological effects following injection of naked DNA. Nature Genet.
P.A. (1996b). Corrective gene transfer in the human skin disorder 10, 161–166.
lamellar ichthyosis. Nature Med. 2, 1263–1267. HENGGE, U.R., WALKER, P.S., and VOGEL, J.C. (1996). Expres-
DALLE, B., PAYEN, E., REGULIER, E., DEGLON, N., ROUYER- sion of naked DNA in human, pig, and mouse skin. J. Clin. Invest.
FESSARD, P., BEUZARD, Y., and AEBISCHER, P. (1999). Im- 97, 2911–2916.
provement of mouse beta-thalassemia upon erythropoietin delivery JENSEN, U.B., JENSEN, T.G., JENSEN, P.K., RYGAARD, J.,
by encapsulated myoblasts. Gene Ther. 6, 157–161. HANSEN, B.S., FOGH, J., KOLVRAA, S., and BOLUND, L.
DAVIDSON, J.M., KRIEG, T., and EMING, S.A. (2000). Particle-me- (1994). Gene transfer into cultured human epidermis and its trans-
diated gene therapy of wounds. Wound Repair Regen. 8, 452–459. plantation onto immunodeficient mice: an experimental model for
DELLAMBRA, E., PELLEGRINI, G., GUERRA, L., FERRARI, G., somatic gene therapy. J. Invest. Dermatol. 103, 391–394.
ZAMBRUNO, G., MAVILIO, F., and DE LUCA, M. (2000). To- KAFRI, T., VAN PRAAG, H., OUYANG, L., GAGE, F.H., and
ward epidermal stem cell-mediated ex vivo gene therapy of junc- VERMA, I.M. (1999). A packaging cell line for lentivirus vectors.
tional epidermolysis bullosa. Hum. Gene Ther. 11, 2283–2287. J. Virol. 73, 576–584.
DENG, H., LIN, Q., and KHAVARI, P.A. (1997). Sustainable cuta- KAY, M.A., MANNO, C.S., RAGNI, M.V., LARSON, P.J., COUTO,
neous gene delivery. Nature Biotechnol. 15, 1388–1391. L.B., MCCLELLAND, A., GLADER, B., CHEW, A.J., TAI, S.J.,
DENG, H., CHOATE, K.A., LIN, Q., and KHAVARI, P.A. (1998). HERZOG, R.W., ARRUDA, V., JOHNSON, F., SCALLAN, C.,
High efficiency gene transfer and pharmacologic selection of genet- SKARSGARD, E., FLAKE, A.W., and HIGH, K.A. (2000). Evi-
ically engineered human keratinocytes. BioTechniques. 25, 274–280. dence for gene transfer and expression of factor IX in haemophilia
DEPOLO, N.J., REED, J.D., SHERIDAN, P.L., TOWNSEND, K., B patients treated with an AAV vector. Nat Genet. 24, 257–61.
SAUTER, S.L., JOLLY, D.J., and DUBENSKY, T.W. (2000). VSV- KHAVARI, P.A. (2000). Genetic correction of inherited epidermal dis-
G pseudotyped lentiviral vector particles produced in human cells orders. Hum. Gene Ther. 11, 2277–2282.
are inactivated by human serum. Mol. Ther. 2, 218–222. LEMIEUX, P., GUERIN, N., PARADIS, G., PROULX, R.,
DOMASHENKO, A., GUPTA, S., and COTSARELIS, G. (2000). Ef- CHISTYAKOVA, L., KABANOV, A., and ALAKHOV, V. (2000).
ficient delivery of transgenes to human hair follicle progenitor cells A combination of poloxamers increases gene expression of plasmid
using topical lipoplex. Nature Biotechnol. 18, 420–423. DNA in skeletal muscle. Gene Ther. 7, 986–991.
1558 BAEK ET AL.

LEVER, A.M., KAYE, J.F., MCCANN, E., CHADWICK, D., DOR- STITZ, J., BUCHHOLZ, C.J., ENGELSTADTER, M., UCKERT, W.,
MAN, N., THOMAS, J., and ZHAO, J. (1999). Lentivirus vectors BLOEMER, U., SCHMITT, I., and CICHUTEK, K. (2000). Lentivi-
for gene therapy. Biochem. Soc. Trans. 27, 841–7. ral vectors pseudotyped with envelope glycoproteins derived from
LI, L., and HOFFMAN, R.M. (1995). The feasibility of targeted se- gibbon ape leukemia virus and murine leukemia virus 10A1. Virol-
lective gene therapy of the hair follicle. Nature Med. 1, 705–706. ogy. 273, 16–20.
LU, B., FEDEROFF, H.J., WANG, Y., GOLDSMITH, L.A., and SVENSSON, E.C., BLACK, H.B., DUGGER, D.L., TRIPATHY, S.K.,
SCOTT, G. (1997). Topical application of viral vectors for epider- GOLDWASSER, E., HAO, Z., CHU, L., and LEIDEN, J.M. (1997).
mal gene transfer. J. Invest. Dermatol. 108, 803–808. Long-term erythropoietin expression in rodents and non-human pri-
MEDALIE, D.A., EMING, S.A., TOMPKINS, R.G., YARMUSH, mates following intramuscular injection of a replication-defective ad-
M.L., KRUEGER, G.G., and MORGAN, J.R. (1996). Evaluation of enoviral vector. Hum. Gene Ther. 8, 1797–1806.
human skin reconstituted from composite grafts of cultured ker- TEUMER, J., LINDAHL, A., and GREEN, H. (1990). Human growth
atinocytes and human acellular dermis transplanted to athymic mice. hormone in the blood of athymic mice grafted with cultures of hor-
J. Invest. Dermatol. 107, 121–127. mone-secreting human keratinocytes. Faseb J. 4, 3245–3250.
MENG, X., SAWAMURA, D., TAMAI, K., HANADA, K., ISHIDA, UITTO, J., and PULKKINEN, L. (2000). The genodermatoses: candi-
H., and HASHIMOTO, I. (1998). Keratinocyte gene therapy for sys- date diseases for gene therapy. Hum. Gene Ther. 11, 2267–2275.
temic diseases. Circulating interleukin 10 released from gene-trans- VOGEL, J.C. (2000). Nonviral skin gene therapy. Hum. Gene Ther.
ferred keratinocytes inhibits contact hypersensitivity at distant areas 11, 2253–2259.
of the skin. J. Clin. Invest. 101, 1462–1467. WANG, C., QUEVEDO, M.E., LANNUTTI, B.J., GORDON, K.B.,
MORGAN, J.R., BARRANDON, Y., GREEN, H., and MULLIGAN, GUO, D., SUN, W., and PALLER, A. S. (1999). In vivo gene ther-
R.C. (1987). Expression of an exogenous growth hormone gene by apy with interleukin-12 inhibits primary vascular tumor growth and
transplantable human epidermal cells. Science. 237, 1476–1479. induces apoptosis in a mouse model. J. Invest. Dermatol. 112, 775–781.
NALDINI, L., BLOMER, U., GALLAY, P., ORY, D., MULLIGAN, WANG, X., ZINKEL, S., POLONSKY, K., and FUCHS, E. (1997).
R., GAGE, F.H., VERMA, I.M., and TRONO, D. (1996). In vivo Transgenic studies with a keratin promoter-driven growth hormone
gene delivery and stable transduction of nondividing cells by a transgene: prospects for gene therapy. Proc. Natl. Acad. Sci. USA
lentiviral vector. Science. 272, 263–267. 94, 219–226.
PETERSEN, M.J., KAPLAN, J., JORGENSEN, C.M., SCHMIDT, YAN, Z., ZHANG, Y., DUAN, D., and ENGELHARDT, J.F. (2000).
L.A., LI, L., MORGAN, J.R., KWAN, M.K., and KRUEGER, G.G. Trans-splicing vectors expand the utility of adeno-associated virus
(1995). Sustained production of human transferrin by transduced fi- for gene therapy. Proc. Natl. Acad. Sci. USA 97, 6716–6721.
broblasts implanted into athymic mice: a model for somatic gene YE, X., RIVERA, V.M., ZOLTICK, P., CERASOLI, F., SCHNELL,
therapy. J. Invest. Dermatol. 104, 171–176. M.A., GAO, G., HUGHES, J.V., GILMAN, M., and WILSON, J.M.
ROBBINS, P.B., LIN, Q., GOODNOUGH, J.B., TIAN, H., CHEN, X., (1999). Regulated delivery of therapeutic proteins after in vivo so-
and KHAVARI, P.A. (2001). In vivo restoration of laminin 5 beta 3 matic cell gene transfer. Science 283, 88–91.
expression and function in junctional epidermolysis bullosa. Proc. ZUFFEREY, R., DULL, T., MANDEL, R.J., BUKOVSKY, A.,
Natl. Acad. Sci. USA 98, 5193–5198. QUIROZ, D., NALDINI, L., and TRONO, D. (1998). Self-inacti-
RUDICH, S.M., ZHOU, S., SRIVASTAVA, R., ESCOBEDO, J.A., vating lentivirus vector for safe and efficient in vivo gene delivery.
PEREZ, R.V., and MANNING, W.C. (2000). Dose response to a sin- J. Virol. 72, 9873–9880.
gle intramuscular injection of recombinant adeno-associated virus-
erythropoietin in monkeys. J. Surg. Res. 90, 102–108. Address reprint requests to:
SEITZ, C.S., GIUDICE, G.J., BALDING, S.D., MARINKOVICH, Dr. Paul A. Khavari
M.P., and KHAVARI, P.A. (1999). BP180 gene delivery in junc- Program in Epithelial Biology
tional epidermolysis bullosa. Gene Ther. 6, 42–47.
269 Campus Drive, Room 2145
SERGUERA, C., BOHL, D., ROLLAND, E., PREVOST, P., and
HEARD, J.M. (1999). Control of erythropoietin secretion by doxy-
Stanford, CA 94305
cycline or mifepristone in mice bearing polymer-encapsulated engi-
neered cells. Hum. Gene Ther. 10, 375–383. E-mail: khavari@CMGM.stanford.edu
SHARKEY, C.M., NORTH, C.L., KUHN, R.J., and SANDERS, D.A.
(2001). Ross River Virus Glycoprotein-Pseudotyped Retroviruses Received for publication March 21, 2001; accepted after revi-
and Stable Cell Lines for Their Production. J. Virol. 75, 2653–2659. sion July 2, 2001.
This article has been cited by:

1. Nicklas Heine Staunstrup, Rasmus O. Bak, Yujia Cai, Lars Svensson, Thomas K. Petersen, Cecilia Rosada,
Karin Stenderup, Lars Bolund, Jacob Giehm Mikkelsen. 2013. A lentiviral vector-based genetic sensor
system for comparative analysis of permeability and activity of vitamin D3 analogues in xenotransplanted
human skin. Experimental Dermatology 22:3, 178-183. [CrossRef]
2. Barbara Geusens, Tine Strobbe, Stefanie Bracke, Peter Dynoodt, Niek Sanders, Mireille Van Gele, Jo
Lambert. 2011. Lipid-mediated gene delivery to the skin. European Journal of Pharmaceutical Sciences 43:4,
199-211. [CrossRef]
3. Matthias Titeux, Alain HovnanianSkin Gene and Cell Therapy 140.1-140.22. [CrossRef]
4. Nikolai Kunicher, Tomer Tzur, Dalit Amar, Malka Chaouat, Barak Yaacov, Amos Panet. 2011.
Characterization of factors that determine lentiviral vector tropism in skin tissue using an ex vivo model.
The Journal of Gene Medicine 13:4, 209-220. [CrossRef]
5. Wolfgang Pfützner. 2010. Vectors for gene therapy of skin diseases. JDDG: Journal der Deutschen
Dermatologischen Gesellschaft 8:8, 582-590. [CrossRef]
6. Frank Scheidemann, Jean-Philippe Therrien, Jon Vogel, Wolfgang Pfützner. 2010. In vivo synthesis
and secretion of erythropoietin by genetically modified primary human keratinocytes grafted onto
immunocompromised mice. Experimental Dermatology 19:3, 289-297. [CrossRef]
7. Maria Jakobsen, Karin Stenderup, Cecilia Rosada, Brian Moldt, Søren Kamp, Tomas N Dam, Thomas G
Jensen, Jacob Giehm Mikkelsen. 2009. Amelioration of Psoriasis by Anti-TNF-α RNAi in the Xenograft
Transplantation Model. Molecular Therapy 17:10, 1743-1753. [CrossRef]
8. Frank Scheidemann, Maria Löser, Andrea Niedermeier, Arno Kromminga, Jean-Philippe Therrien, Jon
Vogel, Wolfgang Pfützner. 2008. The skin as a biofactory for systemic secretion of erythropoietin: potential
of genetically modified keratinocytes and fibroblasts. Experimental Dermatology 17:6, 481-488. [CrossRef]
9. Yasushi Kikuchi, Katsuto Tamai, Yasufumi Kaneda. 2008. Cutaneous gene delivery. Journal of
Dermatological Science 50:2, 87-98. [CrossRef]
10. Nikolai Kunicher, Haya Falk, Barak Yaacov, Tomer Tzur, Amos Panet. 2008. Tropism of Lentiviral Vectors
in Skin Tissue. Human Gene Therapy 19:3, 255-266. [Abstract] [Full Text PDF] [Full Text PDF with
Links]
11. Qian Wang, Heini Ilves, Pauline Chu, Christopher H Contag, Devin Leake, Brian H Johnston, Roger
L Kaspar. 2007. Delivery and Inhibition of Reporter Genes by Small Interfering RNAs in a Mouse Skin
Model. Journal of Investigative Dermatology 127:11, 2577-2584. [CrossRef]
12. A Hachiya, P Sriwiriyanont, A Patel, N Saito, A Ohuchi, T Kitahara, Y Takema, R Tsuboi, R E Boissy,
M O Visscher, W M James, G P Kobinger. 2007. Gene transfer in human skin with different pseudotyped
HIV-based vectors. Gene Therapy 14:8, 648-656. [CrossRef]
13. Martin Laimer, Christoph M Lanschuetzer, Helmut Hintner, Johann W Bauer. 2006. Current approaches
to cutaneous gene therapy. Expert Review of Dermatology 1:6, 833-853. [CrossRef]
14. Paul A. Khavari. 2006. Modelling cancer in human skin tissue. Nature Reviews Cancer 6:4, 270-280.
[CrossRef]
15. Alfred S. Lewin, Peter M. Glazer, Leonard M. Milstone. 2005. Gene Therapy for Autosomal Dominant
Disorders of Keratin. Journal of Investigative Dermatology Symposium Proceedings 10:1, 47-61. [CrossRef]
16. Z. Lu, S. Ghazizadeh. 2005. Host immune responses in ex vivo approaches to cutaneous gene therapy
targeted to keratinocytes. Experimental Dermatology 14:10, 727-735. [CrossRef]
17. U HENGGE, W BARDENHEUER, R DOROUDI, A MIRMOHAMMADSADEGH. 2005. Thérapie
génique et peau. Annales de Dermatologie et de Vénéréologie 132:2, 154-163. [CrossRef]
18. Ulrich R. Hengge. 2005. Progress and prospects of skin gene therapy: a ten year history. Clinics in
Dermatology 23:1, 107-114. [CrossRef]
19. U.R. Hengge, W. Bardenheuer. 2004. Gene therapy and the skin. American Journal of Medical Genetics
131C:1, 93-100. [CrossRef]
20. Junhui Song, Kenji Izumi, Thomas Lanigan, Stephen E. Feinberg. 2004. Development and characterization
of a canine oral mucosa equivalent in a serum-free environment. Journal of Biomedical Materials Research
71A:1, 143-153. [CrossRef]
21. Soosan Ghazizadeh, Anne B. Katz, Robin Harrington, Lorne B. Taichman. 2004. Lentivirus-Mediated
Gene Transfer to Human Epidermis. Journal of Investigative Dermatology Symposium Proceedings 9:3,
269-275. [CrossRef]
22. Thomas G Jensen. 2004. Strategies for long-term gene expression in the skin to treat metabolic disorders.
Expert Opinion on Biological Therapy 4:5, 677-682. [CrossRef]
23. Shariff Dunlap, Xiaobing Yu, Linzhao Cheng, Curt I. Civin, Rhoda M. Alani. 2004. High-Efficiency
Stable Gene Transduction in Primary Human Melanocytes Using a Lentiviral Expression System. Journal
of Investigative Dermatology 122:2, 549-551. [CrossRef]
24. Ricardo Quinonez, Richard E. Sutton. 2002. Lentiviral Vectors for Gene Delivery into Cells. DNA and Cell
Biology 21:12, 937-951. [Abstract] [Full Text PDF] [Full Text PDF with Links]
25. D. Sawamura, M. Akiyama, H. Shimizu. 2002. Direct injection of naked DNA and cytokine transgene
expression: implications for keratinocyte gene therapy. Clinical and Experimental Dermatology 27:6,
480-484. [CrossRef]
26. Tongyu Cao, Sophia Y. Tsai, Bert W. O'Malley, Xiao-Jing Wang, Dennis R. Roop. 2002. The Epidermis
as a Bioreactor: Topically Regulated Cutaneous Delivery into the Circulation. Human Gene Therapy 13:9,
1075-1080. [Abstract] [Full Text PDF] [Full Text PDF with Links]
27. Itaru Suzuki, Sungbin Im, Akihiro Tada, Cathy Scott, Can Akcali, Mary Beth Davis, Greg Barsh, Vincent
Hearing, Zalfa Abdel-Malek. 1999. Participation of the Melanocortin-1 Receptor in the UV Control of
Pigmentation. Journal of Investigative Dermatology Symposium Proceedings 4:1, 29-34. [CrossRef]

You might also like