5
Proc. Nati. Acad. Sci. USA Vol. 91, pp. 2353-2357, March 1994 Genetics In vivo hepatic gene therapy: Complete albeit transient correction of factor IX deficiency in hemophilia B dogs MARK A. KAY*t, CHARLES N. LANDEN*, STEVEN R. ROTHENBERG§, LESLI A. TAYLOR¶, FRANCES LELAND*, SANDRA WIEHLE*, BINGLIANG FANG*, DWIGHT BELLINGERt, MILTON FINEGOLDII, ARTHUR R. THOMPSON**, MARJORIE READS, KENNETH M. BRINKHOUS*, AND SAVIO L. C. Woo* *Howard Hughes Medical Institute, Department of Cell Biology and Molecular Genetics, and Departments of §Pediatric Surgery and IIPathology, Baylor College of Medicine, Houston, TX 77030; Departments of *Pathology and ¶Surgery, University of North Carolina, Chapel Hill, NC 27599; and **Puget Sound Blood Center, 921 Terry Avenue, Seattle, WA 98104 Contributed by Kenneth M. Brinkhous, November 23, 1993 ABSTRACT Hemophilia B is a bleeding disorder caused by mutations in the factor IX gene. The disorder is X-linked recessive with a prevalence of about 1 in 30,000 Caucasian males. Factor IX is naturally synthesized in the liver and secreted into blood. Here we report the construction of recom- binant adenoviral vectors containing the canine factor IX cDNA that are capable of transducing hepatocytes in mice at high efficiencies in vivo without partial hepatectomy. The recombinant viral vector was used to treat hemophilia B dogs by direct vector infusion into the portal vasculature of deficient animals. Plasma factor IX concentrations in the treated hemo- philia B dogs increased from 0 to 300% of the level present in normal dogs, resulting in complete amelioration of the disease as demonstrated by normal blood coagulation and hemostatic measurements. Although plasma factor IX concentration started to decline after a few days, therapeutic levels of factor IX persisted for 1-2 months in the treated animals. The results validate the principle of in ivo hepatic gene delivery to reconstitute the genetic deficiency in a large animal model and suggest that gene therapy is achievable when long-acting vec- tors are developed. Hemophilia B is an X-linked disorder resulting in a deficiency of plasma factor IX, which is normally synthesized and secreted from the liver. The disease affects 1 in 30,000 males (1). Various human protein replacement therapies have sig- nificantly improved the clinical outcome of affected individ- uals, yet there still exists a high degree of morbidity and mortality in part due to previous contamination with human immunodeficiency virus and other hepatitis viruses. The current expense of virus-free factor IX usually limits therapy to treatment of bleeding episodes once they occur. Due to the relatively short half-life of factor IX in the circulation, affected individuals have life-long risks of central nervous system bleeds, chronic arthritis, and other life-threatening hemorrhage. Replacement therapy has improved, but has failed to abrogate, chronic arthropathy, particularly in se- verely affected patients. A number of cell types in different organs have been targeted for somatic gene therapy of hemophilia B, which include fibroblasts, myoblasts, endo- thelial cells, keratinocytes, and hepatocytes (2-8). We have pursued the liver as the target organ because hepatocytes represent the natural site of factor IX synthesis and secretion. We have previously been able to transduce hepatocytes in hemophilia B dogs after partial hepatectomy using a retro- viral vector containing the factor IX cDNA (9). This resulted in long-term expression of about 0.1% of the normal plasma factor IX concentration. To attempt to produce greater levels of recombinant factor IX in animals, we have pursued replication-deficient recombinant adenovirus (10-12) as a gene delivery vector for the treatment of hemophilia B. METHODS Canine Factor IX Adenoviral Vectors. The isolation of a functional canine factor IX cDNA has been described (9). This cDNA was cloned into a Bluescript vector containing either the 540-bp phosphoglycerokinase promoter (13) or the Rous sarcoma virus (RSV) long terminal repeat promoter and the 280-bp bovine growth hormone polyadenylylation site. The expression cassette was cloned into a derivative of the PXCX2 plasmid (14), PXCJL.1 (a gift from Frank Graham), and cotransfected with the pJM17 plasmid (15) into 293 cells (16). Recombinant adenoviral plaques were isolated as de- scribed (15) and analyzed for canine factor IX protein pro- duction in vitro. The recombinant viral particles were grown and purified as described (11) except that after double CsCl banding the virus was extensively dialyzed against 10 mM Tris/1 mM MgCl2, pH 7.4/10%o glycerol. The purified virus was stored in aliquots at -80'C. The virus titer was deter- mined by OD measurements and plaque assay and ranged from 1-3 x 1011 plaque-forming units (pfu) per ml. Animal Studies. All the animal studies were performed in accordance with the institutional guidelines at the Baylor College of Medicine and the University of North Carolina. Hemophilia B dogs were from the Chapel Hill inbred strain (17). Direct in vivo adenoviral-mediated gene transfer into hepatocytes of C57BL/6 mice was as described (10). The placement of an infusion port-catheter into the splenic vein of dogs was described (9). Hemostatic coverage was maintained with multiple infusions of fresh frozen normal dog plasma given immediately before and for 2-5 days after the operation (41.4-68.2 units of factor IX per kg). To demonstrate that the exogenous factor IX had decayed to presurgical levels, the plasma factor IX concentrations were monitored by bioassay and whole-blood clotting times (WBCTs) prior to adenoviral administration (at least 2 weeks). Purified adenovirus was diluted with about 2 volumes of sterile DMEM-H medium and given in two infusions via the infusion port; each infusion lasted about 15-25 min, with a 15-min pause between infu- sions. The total volume infused was about 35-40 ml. Hemostatic Parameters After Adenoviral Gene Therapy. The WBCTs, factor IX bioassay, and nonactivated partial throm- boplastin time (PTT) were performed as described (18-20). A modification of the method of Jim (21) was used for the thrombin clotting time (TCT) test. The ELISA immunoassay (8) for quantitation of factor IX antigen was performed by Abbreviations: RSV, Rous sarcoma virus; PTT, partial thrombo- plastin time; WBCT, whole-blood clotting time; TCT, thrombin clotting time; pfu, plaque-forming units. tPresent address: Markey Molecular Medicine Center, Division of Medical Genetics, Department of Medicine, RG-25, University of Washington, Seattle, WA 98195. 2353 The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. §1734 solely to indicate this fact.

of factor IX deficiency in hemophilia B dogs

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Proc. Nati. Acad. Sci. USAVol. 91, pp. 2353-2357, March 1994Genetics

In vivo hepatic gene therapy: Complete albeit transient correctionof factor IX deficiency in hemophilia B dogsMARK A. KAY*t, CHARLES N. LANDEN*, STEVEN R. ROTHENBERG§, LESLI A. TAYLOR¶, FRANCES LELAND*,SANDRA WIEHLE*, BINGLIANG FANG*, DWIGHT BELLINGERt, MILTON FINEGOLDII, ARTHUR R. THOMPSON**,MARJORIE READS, KENNETH M. BRINKHOUS*, AND SAVIO L. C. Woo**Howard Hughes Medical Institute, Department of Cell Biology and Molecular Genetics, and Departments of §Pediatric Surgery and IIPathology, BaylorCollege of Medicine, Houston, TX 77030; Departments of *Pathology and ¶Surgery, University of North Carolina, Chapel Hill, NC 27599; and**Puget Sound Blood Center, 921 Terry Avenue, Seattle, WA 98104

Contributed by Kenneth M. Brinkhous, November 23, 1993

ABSTRACT Hemophilia B is a bleeding disorder causedby mutations in the factor IX gene. The disorder is X-linkedrecessive with a prevalence of about 1 in 30,000 Caucasianmales. Factor IX is naturally synthesized in the liver andsecreted into blood. Here we report the construction of recom-binant adenoviral vectors containing the canine factor IXcDNA that are capable of transducing hepatocytes in mice athigh efficiencies in vivo without partial hepatectomy. Therecombinant viral vector was used to treat hemophilia B dogsby direct vector infusion into the portal vasculature of deficientanimals. Plasma factor IX concentrations in the treated hemo-philia B dogs increased from 0 to 300% of the level present innormal dogs, resulting in complete amelioration of the diseaseas demonstrated by normal blood coagulation and hemostaticmeasurements. Although plasma factor IX concentrationstarted to decline after a few days, therapeutic levels of factorIX persisted for 1-2 months in the treated animals. The resultsvalidate the principle of in ivo hepatic gene delivery toreconstitute the genetic deficiency in a large animal model andsuggest that gene therapy is achievable when long-acting vec-tors are developed.

Hemophilia B is an X-linked disorder resulting in a deficiencyof plasma factor IX, which is normally synthesized andsecreted from the liver. The disease affects 1 in 30,000 males(1). Various human protein replacement therapies have sig-nificantly improved the clinical outcome of affected individ-uals, yet there still exists a high degree of morbidity andmortality in part due to previous contamination with humanimmunodeficiency virus and other hepatitis viruses. Thecurrent expense of virus-free factor IX usually limits therapyto treatment ofbleeding episodes once they occur. Due to therelatively short half-life of factor IX in the circulation,affected individuals have life-long risks of central nervoussystem bleeds, chronic arthritis, and other life-threateninghemorrhage. Replacement therapy has improved, but hasfailed to abrogate, chronic arthropathy, particularly in se-verely affected patients. A number of cell types in differentorgans have been targeted for somatic gene therapy ofhemophilia B, which include fibroblasts, myoblasts, endo-thelial cells, keratinocytes, and hepatocytes (2-8). We havepursued the liver as the target organ because hepatocytesrepresent the natural site offactor IX synthesis and secretion.We have previously been able to transduce hepatocytes inhemophilia B dogs after partial hepatectomy using a retro-viral vector containing the factor IX cDNA (9). This resultedin long-term expression of about 0.1% of the normal plasmafactor IX concentration. To attempt to produce greater levelsof recombinant factor IX in animals, we have pursued

replication-deficient recombinant adenovirus (10-12) as agene delivery vector for the treatment of hemophilia B.

METHODSCanine Factor IX Adenoviral Vectors. The isolation of a

functional canine factor IX cDNA has been described (9).This cDNA was cloned into a Bluescript vector containingeither the 540-bp phosphoglycerokinase promoter (13) or theRous sarcoma virus (RSV) long terminal repeat promoter andthe 280-bp bovine growth hormone polyadenylylation site.The expression cassette was cloned into a derivative of thePXCX2 plasmid (14), PXCJL.1 (a gift from Frank Graham),and cotransfected with the pJM17 plasmid (15) into 293 cells(16). Recombinant adenoviral plaques were isolated as de-scribed (15) and analyzed for canine factor IX protein pro-duction in vitro. The recombinant viral particles were grownand purified as described (11) except that after double CsClbanding the virus was extensively dialyzed against 10 mMTris/1 mM MgCl2, pH 7.4/10%o glycerol. The purified viruswas stored in aliquots at -80'C. The virus titer was deter-mined by OD measurements and plaque assay and rangedfrom 1-3 x 1011 plaque-forming units (pfu) per ml.Animal Studies. All the animal studies were performed in

accordance with the institutional guidelines at the BaylorCollege of Medicine and the University of North Carolina.Hemophilia B dogs were from the Chapel Hill inbred strain(17). Direct in vivo adenoviral-mediated gene transfer intohepatocytes of C57BL/6 mice was as described (10). Theplacement ofan infusion port-catheter into the splenic vein ofdogs was described (9). Hemostatic coverage was maintainedwith multiple infusions of fresh frozen normal dog plasmagiven immediately before and for 2-5 days after the operation(41.4-68.2 units of factor IX per kg). To demonstrate that theexogenous factor IX had decayed to presurgical levels, theplasma factor IX concentrations were monitored by bioassayand whole-blood clotting times (WBCTs) prior to adenoviraladministration (at least 2 weeks). Purified adenovirus wasdiluted with about 2 volumes of sterile DMEM-H mediumand given in two infusions via the infusion port; each infusionlasted about 15-25 min, with a 15-min pause between infu-sions. The total volume infused was about 35-40 ml.

Hemostatic Parameters After Adenoviral Gene Therapy. TheWBCTs, factor IX bioassay, and nonactivated partial throm-boplastin time (PTT) were performed as described (18-20). Amodification of the method of Jim (21) was used for thethrombin clotting time (TCT) test. The ELISA immunoassay(8) for quantitation of factor IX antigen was performed by

Abbreviations: RSV, Rous sarcoma virus; PTT, partial thrombo-plastin time; WBCT, whole-blood clotting time; TCT, thrombinclotting time; pfu, plaque-forming units.tPresent address: Markey Molecular Medicine Center, Division ofMedical Genetics, Department of Medicine, RG-25, University ofWashington, Seattle, WA 98195.

2353

The publication costs of this article were defrayed in part by page chargepayment. This article must therefore be hereby marked "advertisement"in accordance with 18 U.S.C. §1734 solely to indicate this fact.

Proc. Natl. Acad. Sci. USA 91 (1994)

using a polyclonal rabbit anti-dog factor IX antibody, whichhas no cross-reactivity with mouse factor IX. The primaryantibody was diluted 1:200, and the same antibody was con-jugated to horseradish peroxidase as per the manufacturer'sspecification (Pierce) and used at a 1:25 dilution as the secondantibody. Pooled normal dog plasma was considered to con-tain 11.5 pg of canine factor IX per ml and was used as astandard. The sensitivity of this assay was to 1 ng/ml (9). Thesecondary bleeding time (22) was carried out as described.PCR Quanification of Adenoviral DNA in Tisues. One

microgram of DNA was used per 5S-pi reaction mixture.ExperimentalDNAwas diluted by 10-fold serial dilutions from1 pg to 1 ng. Two pairs of primers were used. The first was inthe RSV promoter (5'-GTAAGGTGGTACGATCGTCGT-GCCT-3'), and the second was in the canine factor IX cDNA(5'-TGCAGTTACCACCCATTTTTCAT-3'), giving rise to aspecific 1-kb product. The second pair of primers were in theRSV promoter (5'-GACTCCTAACCGCGTACA-3') and theadenoviral genome (5'-CTCAATCTGTATCTTCATGC-3'),giving rise to a 280-bp product. Control reactions containingDNA from 1 to 105 viral particle were run in parallel. After 30cycles of amplification, 10 AI was applied to a 2% agarose gel.The intensities ofthe bands in the linear region were comparedto calculate the number of viral particles per microgram ofgenomic DNA. From this, the number of adenoviral genomecopies per diploid cell was estimated.

RESULTSRecombinant adenoviral (Ad) vectors were constructed thatcontain the canine factor IX (cFIX) cDNA under the tran-scriptional control of the phosphoglycerokinase promoter

(PGK) or RSV long terminal repeat promoter (Fig. 1A); thesevectors are referred to as Ad/PGK-cFIX and Ad/RSV-cFIX,respectively. The vectors were shown to transduce cells andproduce canine factor IX in cultured fibroblasts prior to theiruse in animals (data not shown). We have previously deter-mined that 1010 recombinant adenoviral particles can beinfused into the portal vein of a mouse, which resulted in100lo hepatocyte transduction without significant pathologicchanges (10). To test the cFIX vectors for their expression bytransduced hepatocytes in vivo, 1010 purified viral particleswere infused into the portal vasculature of mice. The animalswere periodically bled and serum factor IX antigen concen-trations were determined by ELISA (Fig. 1B). The use oftheAd/RSV-cFIX vector resulted in persistent expression for2-3 months at a serum level that was 10- to 20-fold greaterthan the animals infused with the Ad/PGK-cFIX. Thus theformer vector was used to treat hemophilia B dogs.To determine whether dog hepatocytes could be trans-

duced with adenoviral vectors in vivo, the Ad/RSV-Bgalvector (23) was infused into the portal vasculature of normaldogs via a subcutaneous port with catheter tip placed in thesplenic vein close to itsjunction with the portal vein (9). After8 days of viral transduction the animals were sacrificed, andtissues and hepatocytes were obtained for 5-bromo-4-chloro-3-indolyl ,(D-galactoside (X-gal) staining. The cultured cellswere stained with X-gal within 12 hr of plating. The trans-duction efficiency was determined by counting the propor-tion of blue cells, which ranged from about 20% to 50%obetween different animals. X-gal-stained histochemical liversections showed similar results (data not shown). Routine

A

-. PGK cFIX bpA....I.... E.I.i

36 kb

...-..7. RSV cFiX b AE:-.:B

x

0Cu

ac.'

Ea3C,,

150Days postinfusion

300

FIG. 1. Construction of adenoviral vectors containing the canine factor IX cDNA and direct hepatic gene transfer in mice. (A) The DNAstructure ofthe Ad/PGK-cFIX and Ad/RSV-cFIX adenoviral vectors. The shaded areas represent viral sequences. PGK, phosphoglycerokinasepromoter; bpA, bovine polyadenylylation signal. (B) Serum canine factor IX antigen concentrations in mice after adenoviral-mediated hepaticgene transfer. Recombinant adenovirus (1 x 1010 pfu) particles were infused into the portal vein of C57BL/6. Periodic blood samples wereobtained for canine factor IX analysis by ELISA. Each line represents an individual animal. The open circles represent animals infused withthe Ad/PGK-cFIX vector, and the closed circles represent the animals infused with the Ad/RSV-cFIX vector. Control animals infused withthe same quantity of control adenoviral vectors had no detectable serum canine factor IX antigen (not shown).

2354 Genetics: Kay et al.

Proc. Natl. Acad. Sci. USA 91 (1994) 2355

histological sections of the liver were normal. Occasionalblue cells were found in the spleen but not in other tissues.To determine whether the Ad/RSV-cFIX vector would

correct the coagulation defect in animals, the recombinantvector was injected into the portal vasculature 1 month afterthe installation of infusion ports in two factor IX-deficientdogs from the Chapel Hill inbred strain (17). The missensemutation found in these animals results in the absence ofdetectable factor IX in the plasma (24). Transient palloroccurred during the infusion, and transient temperature ele-vation occurred immediately postinfusion. The temperatureelevation was self-limited and resolved within 8-12 hoursafter treatment. It is not known if this was the result of theadenovirus or a copurifying contaminant.Plasma was obtained periodically for canine factor IX

measurements by immunologic and bioassays. Hemostaticparameters were monitored by changes in the WBCTs andPTTs. The factor IX concentrations in dogs 1 and 2 reachedsupraphysiologic values during the first 4 days after adeno-viral vector infusion: dog 2 had 300% of the normal plasmalevel on day 1, whereas dog 1 had 250%o ofthe normal plasmalevel on day 2 (Fig. 2A). The plasma factor IX levels startedto decline, and by 3 weeks, levels were about 1% of theaverage in normal dogs; by two months they were around0.1% ofnormal and became undetectable in one ofthe treatedanimals after 100 days. The PTT, a measure of the intrinsicclotting mechanism, rapidly shortened from 174-1% sec to47-64 sec by the fifth day (normal range, 42-47 sec) (Fig. 2B).The WBCTs in the treated animals became normal or nearnormal 1-8 days after therapy began (Fig. 2C). Thereafter,both the PTTs and WBCTs slowly increased, which isconsistent with the gradual decline in plasma factor IX levels.

A

E

0

0CoU-

B

0U)n

tL

100,000 -

10,000

1000

100

10

0.1

0.01-10 0

Because hemophilia B dogs 1 and 2 had therapeutic factorIX levels within 24 hr ofinfusion, a third dog was infused withthe Ad/RSV-cFIX adenoviral vector, and plasma was sam-pled every 4-6 hr (Fig. 2D). Normal levels of factor IX wereachieved by 18 hr, and the peak concentration was obtainedat 48 hr, before the slow decline started. In this animal, twoadditional tests were performed prior to and after treatment,TCT and the secondary bleeding time. The TCT was mea-sured to estimate the fibrinogen concentration because of thetransient pallor and febrile reaction the dogs experiencedwith adenoviral gene therapy. The TCT is normal in hemo-philia B dogs; however, it was transiently prolonged afteradenoviral administration. The TCT value at 6 hr was 15.6sec, with the greatest prolongation of 22.2 sec at 12 hr, anda return to the normal value of 12.1 sec at 96 hr. These datasuggest the presence of a transient hypofibrinogenemia. Forthe secondary bleeding time, the pretreatment value was 14.5min. On days 1-8 posttreatment, the values were in the rangeof 1.25-1.75 min, which is normal. Thus, the recombinantfactor IX was able to correct the hemophilic animal's re-sponse to a hemorrhagic challenge.The Ad/RSV-Bgal studies suggest that after portal vein

infusion the majority of virus results in transduction of liver,with small amounts of the virus infecting other tissues. Toquantitate this, a similar dose ofAd/RSV-cFIX virus used totreat the hemophilia B dogs was infused into a female carrier.The hemophilia B heterozygote had a baseline value of 5200ng offactor IX per ml ofplasma; the factor IX level was raisedto 33,000 ng/ml on the third day. The animal was sacrificedat this time, and genomic DNA was extracted to determinethe relative distribution of the recombinant adenoviral ge-nome in tissues by a semiquantitative PCR assay (Table 1).

Days postinfusion

20 40Days postinfusion

40 60Hours postinfusion

FIG. 2. Hemostatic measurements after Ad/RSV-cFIX administration to hemophilia B dogs. Three factor IX-deficient animals (dog 1: 9months old, 14.5 kg; dog 2: 17 months old, 17.3 kg; dog 3: 5.5 months old, 13.9 kg) were infused with Ad/RSV-cFIX (2.4 x 1012 pfu) particlesvia a subcutaneous port. (A-C) Dogs 1 and 2. Circles, hemophilia B dog 1; squares, hemophilia B dog 2. Blood sampling was performedperiodically. (A) Plasma factor IX concentrations. Open symbols, factor IX measurements as determined by a biological assay; solid symbols,factor IX as determined by an immunologic ELISA assay. (B) PTTs. (C) WBCTs. (D) Plasma factor IX concentations for dog 3. The valuesobtained on day 0 were collected prior to any experimental procedure.

Genetics: Kay et al.

Proc. Natl. Acad. Sci. USA 91 (1994)

The amount of adenoviral DNA per cell averaged 0.7-7copies per hepatocyte, which is at least 10-fold greater thanthe next abundant tissues, which included the spleen andstomach. The presence ofsome adenovirus in these tissues isnot unexpected because of the vascular tributaries of thesplenic vein from these organs. Other organs listed in thetable had significantly less adenoviral DNA.At the time of sacrifice, tissues were also prepared for

routine histological analysis. The results showed some milddegeneration and necrosis that was most apparent in the rightlateral lobe of the liver. Transient mild hepatic necrosis hasbeen observed in mice transduced with adenoviral vectors(10). It is of interest to note that in the Ad/RSV-Bgal-infuseddogs, there was no apparent liver pathology 8 days after viralinfusion. Other organs that were studied histologically in-cluded the kidney, thymus, lymph nodes, duodenum, spleen,brain, stomach, colon, heart, lung, and thyroid, which wereall normal.

DISCUSSIONRecombinant adenoviral vectors have been used to transfergenes into a number of cell targets. We and others have usedthese vectors for the direct in vivo transfer of genes into theliver of animals (10-12). The study here demonstrates thatrecombinant adenoviral vectors can be used in a large animalmodel to completely ameliorate the coagulation defect inhemophilia B that results from the synthesis of high levels offactor IX in the liver. We have demonstrated in a rodentmodel that the degree of hepatic transduction is directlyrelated to the dose of adenovirus infused. Because theamount of plasma factor IX produced is greater than thenormal levels, less adenovirus will be needed to achieve thewild-type plasma concentration. In fact, considerably smallerdoses will be needed to achieve 25% reconstitution, which isconsidered curative. One animal studied did have mild livernecrosis with adenoviral infusion. Transient mild hepaticnecrosis has been demonstrated in the mouse in a dose-dependent manner (10). Thus, it is likely that the lower doseofadenovirus required for curative levels offactor IX will notresult in hepatic toxicity.The animals treated with adenovirus demonstrated a lag

from the period of normal factor IX concentrations andnormalization of the PTT. This in combination with thetransient febrile reaction led us to consider the possibility ofa mild disseminated intravascular coagulation event. Thishypothesis is supported by the transient hypofibrinogenemiademonstrated in dog 3. Whether this event is a direct cause

Table 1. Tissue distribution of adenoviral DNA in a hemophiliaB heterozygote

Tissue Copies per ug of DNA Copies per cellLiver 105-106 0.7-7Spleen 104-105 0.07-0.7Stomach 104-105 0.07-0.7Lung 103-104 0.007-0.07Kidney 103-104 0.007-0.07Heart 103-104 0.007-0.07Ovary 103-104 0.007-0.07Muscle 103-104 0.007-0.07Lymph node 102-103 0.0007-0.007Brain 102-103 0.0007-0.007A 4.5-month-old factor IX-deficient heterozygous female dog (14

kg) was infused with 3 x 1012 Ad/RSV-cFIX adenoviral particles viathe portal vein. Peak factor IX plasma levels were similar to thoseobtainedfor dogs 1-3. Three days later the-animal was sacrificed, andtissues obtained for DNA preparation and a semiquantitative PCRassay were used to estimate the amount of recombinant adenoviralDNA in each tissue.

ofadenovirus or a copurifying contaminant is not known. Wehave previously demonstrated a similar clinical response to achallenge of minute quantities of fetal calf serum (25), whichis used to propagate the adenovirus, but until further studiesare carried out, the cause ofthe transient clinical reaction andhypofibrinogenemia cannot be determined. It is likely that thereaction will be less significant if lower doses of adenovirusare administered to these animals.Expression after adenoviral-mediated gene transfer in the

hemophilia B dogs is transient and most likely results fromthe loss of DNA from transduced cells as we have demon-strated in the mouse (10). Interestingly, factor IX expressionpersisted longer in the mouse than the dog for reasons that areunclear at this time, although the canine factor IX concen-trations did eventually begin to fall in the mouse. The kineticsof factor IX expression is different in mice infused with theAd/PGK-cFIX vector than with the Ad/RSV-cFIX vector.The former vector results in peak expression within the firstweek followed by a slow decline. The mice transduced withthe Ad/RSV-cFIX vector reach peak serum factor IX con-centrations between 2 and 4 weeks after infusion before theconcentrations begin to decline. In contrast, when this vectorwas used in the hemophilia B dogs, the maximal concentra-tions of plasma factor IX were detected within the first 2days. The differences in expression may in part be due todifferences in gene transcription from this promoter in thetwo species. The fall in serum factor IX in mice is consistentwith the decline in the percentage of(-galactosidase-positivehepatocytes that were found at various times after hepaticAd/RSV-3gal administration in our earlier studies (10).The nature of adenoviral DNA loss from transduced cells

is currently not known. It is possible that transduced cells areslowly replaced or that the episomalDNA is slowly degradedin hepatocytes. We have attempted to infuse a second doseof recombinant adenovirus in one hemophilia B animal afterrecombinant factor IX was no longer detectable. No recom-binant factor IX was present in the plasma after reinfusion,suggesting an immunologic block to reinfection (data notshown). Antibodies to fiber are believed to be neutralizing,and the presence of antibodies may be responsible for theinability to reinfuse the animals and get secondary genetransfer. Additional studies will be necessary to define animmunologic block and to establish a causal relationship withthe inability to give repeated doses for the purposes ofhepaticgene therapy for hemophilia B.The high efficiency ofgene transfer and expression into the

liver, albeit transient, demonstrates that in vivo gene deliveryto the liver can be practical in humans in the future. Manymetabolic disorders can be improved with even transientgene expression in vivo. This makes the recombinant adeno-viral vector system an attractive one for future hepatic genetransfer and therapy, particularly if the lack of persistencecan be addressed by further vector development.

We thank Robin Raymer and Pam McElveen (Chapel Hill, NC) fortheir expert technical assistance. This work was supported in part byNational Institutes of Health Grants DK 44080 (Baylor College ofMedicine) and HL 01648-46 and HL 26309-12 (University of NorthCarolina, Chapel Hill). M.A.K. is a recipient of an individualNational Research Service Award Fellowship AwardGM 13894, andS.L.C.W. is an Investigator ofthe Howard Hughes Medical Institute.

1. Roberts, H. R. & Lozier, J. N. (1991) in Clinical Aspects andTherapyfor HemophiliaB in Hematology: Basic Principles andPractice, eds. Hoffman, R., Benz, E., Jr., Shattil, S., Furie, B.& Cohen, H. (Churchill Livingstone, New York), pp. 1325-1331.

2. Brinkhous, K. M. (1992) Thromb. Res. 67, 329-338.3. Thompson, A. R., Palmer, T. D., Lynch, C. M. & Miller,

A. D. (1991) Curr. Stud. Hematol. Blood Transfus. 58, 59-62.

2356 Genetics: Kay et al.

Proc. Natl. Acad. Sci. USA 91 (1994) 2357

4. Dai, Y., Roman, M., Naviaux, R. K. & Verma, I. (1992) Proc.NatI. Acad. Sci. USA 89, 10892-10895.

5. Miyanohara, A., Johnson, P. A., Elam, R. L., Dai, Y., Witz-tum, J. L., Verma, I. & Freidman, T. (1992) New Biol. 4,238-246.

6. Yao, S. N. & Kurachi, K. (1992) Proc. Natl. Acad. Sci. USA89, 3357-3361.

7. Gerrard, A. J., Hudson, D. L., Brownlee, G. G. & Watt, F. M.(1993) Nature Genet. 3, 180-183.

8. Armentano, D., Thompson, A. R., Darlington, G. & Woo,S. L. C. (1990) Proc. Nati. Acad. Sci. USA 87, 6141-6145.

9. Kay, M. A., Rothenberg, S., Landen, C. N., Bellinger, D. A.,Leland, F., Toman, C., Finegold, M., Thompson, A. R., Read,M. S., Brinkhous, K. M. & Woo, S. L. C. (1993) Science 262,117-119.

10. Li, Q. T., Kay, M. A., Finegold, M., Stratford-Perricaudet,L. D. & Woo, S. L. C. (1993) Hum. Gene Ther. 4, 403-409.

11. Herz, J. & Gerard, R. (1993) Proc. NatI. Acad. Sci. USA 90,2812-2816.

12. Jaffe, H. A., Danel, C., Longenecker, G., Metzger, M., Se-toguchi, Y., Rosenfeld, M. A., Gant, T. W., Thorgensson,S. S., Stratford-Perricaudet, L. D., Perricaudet, M., Pavrant,A., Lecocq, J. P. & Crystal, R. G. (1992) Nature Genet. 1,372-378.

13. Soriano, P., Friedrich, G. & Lawinger, P. (1991) J. Virol. 65,2314-2319.

14. Spessot, R., Inchley, K., Hupel, T. M. & Baccheti, S. (1989)Virology 168, 378-387.

15. McGrory, W. J., Bautista, D. S. & Graham, F. L. (1988)Virology 163, 614-617.

16. Graham, F. L. & Ludvik, P. (1991) Methods Mol. Biol. 7,109-127.

17. Brinkhous, K. M., Davis, P. D., Garris, J. B., Diness, V. &Read, M. S. (1973) Blood 41, 577-585.

18. Brinkhous, K. M., Landen, C. N. & Read, M. S. (1993) Blood82, Suppl. 1, 592 (abstr.).

19. Brinkhous, K. M., Hedner, U., Garris, J. B., Diness, V. &Read, M. S. (1989) Proc. Natl. Acad. Sci. USA 86, 1382-1386.

20. Langdell, R. D., Wagner, R. H. & Brinkhous, K. M. (1953) J.Lab. Clin. Med. 41, 637-647.

21. Jim, R. T. S. (1957) J. Lab. Clin. Med. 50, 45-60.22. Brinkhous, K. M., Sandberg, H., Garris, J. B., Mattsson, C.,

Palm, M., Griggs, T. & Read, M. S. (1985) Proc. Natl. Acad.Sci. USA 82, 8752-8756.

23. Stratford-Perricaudet, L. D., Makeh, I., Perricaudet, M. &Briand, P. (1992) J. Clin. Invest. 90, 626-630.

24. Evans, J. P., Brinkhous, K. M., Brager, G. D., Reisner, H. M.& High, K. A. (1989) Proc. NatI. Acad. Sci. USA 86, 10095-10098.

25. Kay, M. A., Baley, P., Rothenberg, S., Leland, F., Fleming,L., Parker-Ponder, K., Liu, T. J., Finegold, M., Darlington,G., Pokorny, W. & Woo, S. L. C. (1992) Proc. NatI. Acad. Sci.USA 89, 89-93.

Genetics: Kay et al.