Drug/drug delivery systems for the prevention and treatment of vascular disease

Abstract
A drug and drug delivery system may be utilized in the treatment of vascular disease. A local delivery system is coated with rapamycin or other suitable drug, agent or compound and delivered intraluminally for the treatment and prevention of neointimal hyperplasia following percutaneous transluminal coronary angiography. The local delivery of the drugs or agents provides for increased effectiveness and lower systemic toxicity.
Description
BACKGROUND OF THE INVENTION

1. Field of the Invention


The present invention relates to drugs and drug delivery systems for the prevention and treatment of vascular disease, and more particularly to drugs and drug delivery systems for the prevention and treatment of neointimal hyperplasia.


2. Discussion of the Related Art


Many individuals suffer from circulatory disease caused by a progressive blockage of the blood vessels that perfuse the heart and other major organs with nutrients. More severe blockage of blood vessels in such individuals often leads to hypertension, ischemic injury, stroke, or myocardial infarction. Atherosclerotic lesions, which limit or obstruct coronary blood flow, are the major cause of ischemic heart disease. Percutaneous transluminal coronary angioplasty is a medical procedure whose purpose is to increase blood flow through an artery. Percutaneous transluminal coronary angioplasty is the predominant treatment for coronary vessel stenosis. The increasing use of this procedure is attributable to its relatively high success rate and its minimal invasiveness compared with coronary bypass surgery. A limitation associated with percutaneous transluminal coronary angioplasty is the abrupt closure of the vessel which may occur immediately after the procedure and restenosis which occurs gradually following the procedure. Additionally, restenosis is a chronic problem in patients who have undergone saphenous vein bypass grafting. The mechanism of acute occlusion appears to involve several factors and may result from vascular recoil with resultant closure of the artery and/or deposition of blood platelets and fibrin along the damaged length of the newly opened blood vessel.


Restenosis after percutaneous transluminal coronary angioplasty is a more gradual process initiated by vascular injury. Multiple processes, including thrombosis, inflammation, growth factor and cytokine release, cell proliferation, cell migration and extracellular matrix synthesis each contribute to the restenotic process.


While the exact mechanism of restenosis is not completely understood, the general aspects of the restenosis process have been identified. In the normal arterial wall, smooth muscle cells proliferate at a low rate, approximately less than 0.1 percent per day. Smooth muscle cells in the vessel walls exist in a contractile phenotype characterized by eighty to ninety percent of the cell cytoplasmic volume occupied with the contractile apparatus. Endoplasmic reticulum, Golgi, and free ribosomes are few and are located in the perinuclear region. Extracellular matrix surrounds the smooth muscle cells and is rich in heparin-like glycosylaminoglycans which are believed to be responsible for maintaining smooth muscle cells in the contractile phenotypic state (Campbell and Campbell, 1985).


Upon pressure expansion of an intracoronary balloon catheter during angioplasty, smooth muscle cells within the vessel wall become injured, initiating a thrombotic and inflammatory response. Cell derived growth factors such as platelet derived growth factor, fibroblast growth factor, epidermal growth factor, thrombin, etc., released from platelets, invading macrophages and/or leukocytes, or directly from the smooth muscle cells provoke proliferative and migratory responses in medial smooth muscle cells. These cells undergo a change from the contractile phenotype to a synthetic phenotype characterized by only a few contractile filament bundles, extensive rough endoplasmic reticulum, Golgi and free ribosomes. Proliferation/migration usually begins within one to two days post-injury and peaks several days thereafter (Campbell and Campbell, 1987; Clowes and Schwartz, 1985).


Daughter cells migrate to the intimal layer of arterial smooth muscle and continue to proliferate and secrete significant amounts of extracellular matrix proteins. Proliferation, migration and extracellular matrix synthesis continue until the damaged endothelial layer is repaired at which time proliferation slows within the intima, usually within seven to fourteen days post-injury. The newly formed tissue is called neointima. The further vascular narrowing that occurs over the next three to six months is due primarily to negative or constrictive remodeling.


Simultaneous with local proliferation and migration, inflammatory cells invade the site of vascular injury. Within three to seven days post-injury, inflammatory cells have migrated to the deeper layers of the vessel wall. In animal models employing either balloon injury or stent implantation, inflammatory cells may persist at the site of vascular injury for at least thirty days (Tanaka et al., 1993; Edelman et al., 1998). Inflammatory cells therefore are present and may contribute to both the acute and chronic phases of restenosis.


Numerous agents have been examined for presumed anti-proliferative actions in restenosis and have shown some activity in experimental animal models. Some of the agents which have been shown to successfully reduce the extent of intimal hyperplasia in animal models include: heparin and heparin fragments (Clowes, A. W. and Karnovsky M., Nature 265: 25-26, 1977; Guyton, J. R. et al., Circ. Res., 46: 625-634, 1980; Clowes, A. W. and Clowes, M. M., Lab. Invest. 52: 611-616, 1985; Clowes, A. W. and Clowes, M. M., Circ. Res. 58: 839-845, 1986; Majesky et al., Circ. Res. 61: 296-300, 1987; Snow et al., Am. J. Pathol. 137: 313-330, 1990; Okada, T. et al., Neurosurgery 25: 92-98, 1989), colchicine (Currier, J. W. et al., Circ. 80: 11-66, 1989), taxol (Sollot, S. J. et al., J. Clin. Invest. 95: 1869-1876, 1995), angiotensin converting enzyme (ACE) inhibitors (Powell, J. S. et al., Science, 245: 186-188, 1989), angiopeptin (Lundergan, C. F. et al. Am. J. Cardiol. 17(Suppl. B):132B-136B, 1991), cyclosporin A (Jonasson, L. et al., Proc. Natl., Acad. Sci., 85: 2303, 1988), goat-anti-rabbit PDGF antibody (Ferns, G. A. A., et al., Science 253: 1129-1132, 1991), terbinafine (Nemecek, G. M. et al., J. Pharmacol. Exp. Thera. 248: 1167-1174, 1989), trapidil (Liu, M. W. et al., Circ. 81: 1089-1093, 1990), tranilast (Fukuyama, J. et al., Eur. J. Pharmacol. 318: 327-332, 1996), interferon-gamma (Hansson, G. K. and Holm, J., Circ. 84:1266-1272, 1991), rapamycin (Marx, S. O. et al., Circ. Res. 76: 412-417, 1995), corticosteroids (Colburn, M. D. et al., J. Vasc. Surg. 15: 510-518, 1992), see also Berk, B. C. et al., J. Am. Coll. Cardiol. 17: 111B-117B, 1991), ionizing radiation (Weinberger, J. et al., Int. J. Rad. Onc. Biol. Phys. 36: 767-775, 1996), fusion toxins (Farb, A. et al., Circ. Res. 80: 542-550, 1997) antisense oligonucleotides (Simons, M. et al., Nature 359: 67-70, 1992) and gene vectors (Chang, M. W. et al., J. Clin. Invest. 96: 2260-2268, 1995). Anti-proliferative effects on smooth muscle cells in vitro have been demonstrated for many of these agents, including heparin and heparin conjugates, taxol, tranilast, colchicine, ACE inhibitors, fusion toxins, antisense oligonucleotides, rapamycin and ionizing radiation. Thus, agents with diverse mechanisms of smooth muscle cell inhibition may have therapeutic utility in reducing intimal hyperplasia.


However, in contrast to animal models, attempts in human angioplasty patients to prevent restenosis by systemic pharmacologic means have thus far been unsuccessful. Neither aspirin-dipyridamole, ticlopidine, anti-coagulant therapy (acute heparin, chronic warfarin, hirudin or hirulog), thromboxane receptor antagonism nor steroids have been effective in preventing restenosis, although platelet inhibitors have been effective in preventing acute reocclusion after angioplasty (Mak and Topol, 1997; Lang et al., 1991; Popma et al., 1991). The platelet GP IIb/IIIa receptor, antagonist, Reopro is still under study but has not shown promising results for the reduction in restenosis following angioplasty and stenting. Other agents, which have also been unsuccessful in the prevention of restenosis, include the calcium channel antagonists, prostacyclin mimetics, angiotensin converting enzyme inhibitors, serotonin receptor antagonists, and anti-proliferative agents. These agents must be given systemically, however, and attainment of a therapeutically effective dose may not be possible; anti-proliferative (or anti-restenosis) concentrations may exceed the known toxic concentrations of these agents so that levels sufficient to produce smooth muscle inhibition may not be reached (Mak and Topol, 1997; Lang et al., 1991; Popma et al., 1991).


Additional clinical trials in which the effectiveness for preventing restenosis utilizing dietary fish oil supplements or cholesterol lowering agents has been examined showing either conflicting or negative results so that no pharmacological agents are as yet clinically available to prevent post-angioplasty restenosis (Mak and Topol, 1997; Franklin and Faxon, 1993: Serruys, P. W. et al., 1993). Recent observations suggest that the antilipid/antioxidant agent, probucol may be useful in preventing restenosis but this work requires confirmation (Tardif et al., 1997; Yokoi, et al., 1997). Probucol is presently not approved for use in the United States and a thirty-day pretreatment period would preclude its use in emergency angioplasty. Additionally, the application of ionizing radiation has shown significant promise in reducing or preventing restenosis after angioplasty in patients with stents (Teirstein et al., 1997). Currently, however, the most effective treatments for restenosis are repeat angioplasty, atherectomy or coronary artery bypass grafting, because no therapeutic agents currently have Food and Drug Administration approval for use for the prevention of post-angioplasty restenosis.


Unlike systemic pharmacologic therapy, stents have proven effective in significantly reducing restenosis. Typically, stents are balloon-expandable slotted metal tubes (usually, but not limited to, stainless steel), which, when expanded within the lumen of an angioplastied coronary artery, provide structural support through rigid scaffolding to the arterial wall. This support is helpful in maintaining vessel lumen patency. In two randomized clinical trials, stents increased angiographic success after percutaneous transluminal coronary angioplasty, by increasing minimal lumen diameter and reducing, but not eliminating, the incidence of restenosis at six months (Serruys et al., 1994; Fischman et al., 1994).


Additionally, the heparin coating of stents appears to have the added benefit of producing a reduction in sub-acute thrombosis after stent implantation (Serruys et al., 1996). Thus, sustained mechanical expansion of a stenosed coronary artery with a stent has been shown to provide some measure of restenosis prevention, and the coating of stents with heparin has demonstrated both the feasibility and the clinical usefulness of delivering drugs locally, at the site of injured tissue.


Accordingly, there exists a need for effective drugs and drug delivery systems for the effective prevention and treatment of neointimal thickening that occurs after percutaneous transluminal coronary angioplasty and stent implantation.


SUMMARY OF THE INVENTION

The drugs and drug delivery systems of the present invention provide a means for overcoming the difficulties associated with the methods and devices currently in use as briefly described above.


In accordance with one aspect, the present invention is directed to a method for the treatment of intimal hyperplasia in vessel walls. The method comprises the controlled delivery, by release from an intraluminal medical device, of cell cycle inhibitors that act selectively at the G1 phase of the cell cycle.


In accordance with another aspect, the present invention is directed to a drug delivery device. The drug delivery device comprises an intraluminal medical device and a therapeutic dosage of an agent releasably affixed to the intraluminal medical device for the treatment of intimal hyperplasia, constrictive vascular remodeling and inflammation caused by injury.


The drugs and drug delivery systems of the present invention utilize a stent or graft in combination with rapamycin or other drugs/agents/compounds to prevent and treat neointimal hyperplasia, i.e. restenosis, following percutaneous transluminal coronary angioplasty and stent implantation. It has been determined that rapamycin functions to inhibit smooth muscle cell proliferation through a number of mechanisms. It has also been determined that rapamycin eluting stent coatings produce superior effects in humans, when compared to animals, with respect to the magnitude and duration of the reduction in neointimal hyperplasia. Rapamycin administration from a local delivery platform also produces an anti-inflammatory effect in the vessel wall that is distinct from and complimentary to its smooth muscle cell anti-proliferative effect. In addition, it has also been demonstrated that rapamycin inhibits constrictive vascular remodeling in humans.


Other drugs, agents or compounds which mimic certain actions of rapamycin may also be utilized in combination with local delivery systems or platforms.


The local administration of drugs, agents or compounds to stented vessels have the additional therapeutic benefit of higher tissue concentration than that which would be achievable through the systemic administration of the same drugs, agents or compounds. Other benefits include reduced systemic toxicity, single treatment, and ease of administration. An additional benefit of a local delivery device and drug, agent or compound therapy may be to reduce the dose of the therapeutic drugs, agents or compounds and thus limit their toxicity, while still achieving a reduction in restenosis.





BRIEF DESCRIPTION OF THE DRAWINGS

The foregoing and other features and advantages of the invention will be apparent from the following, more particular description of preferred embodiments of the invention, as illustrated in the accompanying drawings.



FIG. 1 is a chart indicating the effectiveness of rapamycin as an anti-inflammatory relative to other anti-inflammatories.



FIG. 2 is a view along the length of a stent (ends not shown) prior to expansion showing the exterior surface of the stent and the characteristic banding pattern.



FIG. 3 is a perspective view of the stent of FIG. 1 having reservoirs in accordance with the present invention.





DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS

As stated above, the proliferation of vascular smooth muscle cells in response to mitogenic stimuli that are released during balloon angioplasty and stent implantation is the primary cause of neointimal hyperplasia. Excessive neointimal hyperplasia can often lead to impairment of blood flow, cardiac ischemia and the need for a repeat intervention in selected patients in high risk treatment groups. Yet repeat revascularization incurs risk of patient morbidity and mortality while adding significantly to the cost of health care. Given the widespread use of stents in interventional practice, there is a clear need for safe and effective inhibitors of neointimal hyperplasia.


Rapamycin is a macroyclic triene antibiotic produced by streptomyces hygroscopicus as disclosed in U.S. Pat. No. 3,929,992. It has been found that rapamycin inhibits the proliferation of vascular smooth muscle cells in vivo. Accordingly, rapamycin may be utilized in treating intimal smooth muscle cell hyperplasia, restenosis and vascular occlusion in a mammal, particularly following either biologically or mechanically mediated vascular injury, or under conditions that would predispose a mammal to suffering such a vascular injury. Rapamycin functions to inhibit smooth muscle cell proliferation and does not interfere with the re-endothelialization of the vessel walls.


Rapamycin functions to inhibit smooth muscle cell proliferation through a number of mechanisms. In addition, rapamycin reduces the other effects caused by vascular injury, for example, inflammation. The operation and various functions of rapamycin are described in detail below. Rapamycin as used throughout this application shall include rapamycin, rapamycin analogs, derivatives and congeners that bind FKBP12 and possess the same pharmacologic properties as rapamycin.


Rapamycin reduces vascular hyperplasia by antagonizing smooth muscle proliferation in response to mitogenic signals that are released during angioplasty. Inhibition of growth factor and cytokine mediated smooth muscle proliferation at the late G1 phase of the cell cycle is believed to be the dominant mechanism of action of rapamycin. However, rapamycin is also known to prevent T-cell proliferation and differentiation when administered systemically. This is the basis for its immunosuppresive activity and its ability to prevent graft rejection.


The molecular events that are responsible for the actions of rapamycin, a known anti-proliferative, which acts to reduce the magnitude and duration of neointimal hyperplasia, are still being elucidated. It is known, however, that rapamycin enters cells and binds to a high-affinity cytosolic protein called FKBP12. The complex of rapamycin and FKPB12 in turn binds to and inhibits a phosphoinositide (PI)-3 kinase called the “mammalian Target of Rapamycin” or TOR. TOR is a protein kinase that plays a key role in mediating the downstream signaling events associated with mitogenic growth factors and cytokines in smooth muscle cells and T lymphocytes. These events include phosphorylation of p27, phosphorylation of p70 s6 kinase and phosphorylation of 4BP-1, an important regulator of protein translation.


It is recognized that rapamycin reduces restenosis by inhibiting neointimal hyperplasia. However, there is evidence that rapamycin may also inhibit the other major component of restenosis, namely, negative remodeling. Remodeling is a process whose mechanism is not clearly understood but which results in shrinkage of the external elastic lamina and reduction in lumenal area over time, generally a period of approximately three to six months in humans.


Negative or constrictive vascular remodeling may be quantified angiographically as the percent diameter stenosis at the lesion site where there is no stent to obstruct the process. If late lumen loss is abolished in-lesion, it may be inferred that negative remodeling has been inhibited. Another method of determining the degree of remodeling involves measuring in-lesion external elastic lamina area using intravascular ultrasound (IVUS). Intravascular ultrasound is a technique that can image the external elastic lamina as well as the vascular lumen. Changes in the external elastic lamina proximal and distal to the stent from the post-procedural timepoint to four-month and twelve-month follow-ups are reflective of remodeling changes.


Evidence that rapamycin exerts an effect on remodeling comes from human implant studies with rapamycin coated stents showing a very low degree of restenosis in-lesion as well as in-stent. In-lesion parameters are usually measured approximately five millimeters on either side of the stent i.e. proximal and distal. Since the stent is not present to control remodeling in these zones which are still affected by balloon expansion, it may be inferred that rapamycin is preventing vascular remodeling.


The data in Table 1 below illustrate that in-lesion percent diameter stenosis remains low in the rapamycin treated groups, even at twelve months. Accordingly, these results support the hypothesis that rapamycin reduces remodeling.


Angiographic In-Lesion Percent Diameter Stenosis (%, Mean±SD and “n=”) in Patients Who Received a Rapamycin-Coated Stent











TABLE 1.0





Coating
Post
4-6 month
12 month


Group
Placement
Follow Up
Follow Up







Brazil
10.6 ± 5.7 (30)
13.6 ± 8.6 (30)
22.3 ± 7.2 (15)


Netherlands
14.7 ± 8.8
22.4 ± 6.4










Additional evidence supporting a reduction in negative remodeling with rapamycin comes from intravascular ultrasound data that was obtained from a first-in-man clinical program as illustrated in Table 2 below.


Matched IVUS Data in Patients Who Received a Rapamycin-Coated Stent











TABLE 2.0







4-Month
12-Month




Follow-Up
Follow-Up


IVUS Parameter
Post (n =)
(n =)
(n =)







Mean proximal vessel area
16.53 ± 3.53
16.31 ± 4.36
13.96 ± 2.26


(mm2)
(27)
(28)
(13)


Mean distal vessel area
13.12 ± 3.68
13.53 ± 4.17
12.49 ± 3.25


(mm2)
(26)
(26)
(14)









The data illustrated that there is minimal loss of vessel area proximally or distally which indicates that inhibition of negative remodeling has occurred in vessels treated with rapamycin-coated stents.


Other than the stent itself, there have been no effective solutions to the problem of vascular remodeling. Accordingly, rapamycin may represent a biological approach to controlling the vascular remodeling phenomenon.


It may be hypothesized that rapamycin acts to reduce negative remodeling in several ways. By specifically blocking the proliferation of fibroblasts in the vascular wall in response to injury, rapamycin may reduce the formation of vascular scar tissue. Rapamycin may also affect the translation of key proteins involved in collagen formation or metabolism.


Rapamycin used in this context includes rapamycin and all analogs, derivatives and congeners that bind FKBP12 and possess the same pharmacologic properties as rapamycin.


In a preferred embodiment, the rapamycin is delivered by a local delivery device to control negative remodeling of an arterial segment after balloon angioplasty as a means of reducing or preventing restenosis. While any delivery device may be utilized, it is preferred that the delivery device comprises a stent that includes a coating or sheath which elutes or releases rapamycin. The delivery system for such a device may comprise a local infusion catheter that delivers rapamycin at a rate controlled by the administrator.


Rapamycin may also be delivered systemically using an oral dosage form or a chronic injectible depot form or a patch to deliver rapamycin for a period ranging from about seven to forty-five days to achieve vascular tissue levels that are sufficient to inhibit negative remodeling. Such treatment is to be used to reduce or prevent restenosis when administered several days prior to elective angioplasty with or without a stent.


Data generated in porcine and rabbit models show that the release of rapamycin into the vascular wall from a nonerodible polymeric stent coating in a range of doses (35-430 ug/15-18 mm coronary stent) produces a peak fifty to fifty-five percent reduction in neointimal hyperplasia as set forth in Table 3 below. This reduction, which is maximal at about twenty-eight to thirty days, is typically not sustained in the range of ninety to one hundred eighty days in the porcine model as set forth in Table 4 below.









TABLE 3.0







Animal Studies with Rapamycin-coated stents.


Values are mean ± Standared Error of Mean










Neointimal




Area
% Change From














Study
Duration
Stent1
Rapamycin
N
(mm2)
Polyme
Metal











Porcine
















98009
14 days
Metal


8
2.04 ± 0.17






1X + rapamycin
153
μg
8
1.66 ± 0.17*
−42%
−19%




1X + TC300 + rapamycin
155
μg
8
1.51 ± 0.19*
−47%
−26%


99005
28 days
Metal


10
2.29 ± 0.21









9
3.91 ± 0.60**






1X + TC30 + rapamycin
130
μg
8
2.81 ± 0.34

+23%




1X + TC100 + rapamycin
120
μg
9
2.62 ± 0.21

+14%


99006
28 days
Metal


12
4.57 ± 0.46






EVA/BMA 3X


12
5.02 ± 0.62

+10%




1X + rapamycin
125
μg
11
2.84 ± 0.31* **
−43%
−38%




3X + rapamycin
430
μg
12
3.06 ± 0.17* **
−39%
−33%




3X + rapamycin
157
μg
12
2.77 ± 0.41* **
−45%
−39%


99011
28 days
Metal


11
3.09 ± 0.27









11
4.52 ± 0.37






1X + rapamycin
189
μg
14
3.05 ± 0.35

 −1%




3X + rapamycin/dex
182/363
μg
14
2.72 ± 0.71

−12%


99021
60 days
Metal


12
2.14 ± 0.25






1X + rapamycin
181
μg
12
2.95 ± 0.38

+38%


99034
28 days
Metal


8
5.24 ± 0.58






1X + rapamycin
186
μg
8
2.47 ± 0.33**

−53%




3X + rapamycin/dex
185/369
μg
6
2.42 ± 0.64**

−54%


20001
28 days
Metal


6
1.81 ± 0.09






1X + rapamycin
172
μg
5
1.66 ± 0.44

 −8%


20007











30 days
Metal


9
2.94 ± 0.43






1XTC + rapamycin
155
μg
10
1.40 ± 0.11*

−52%*








Rabbit
















99019
28 days
Metal


8
1.20 ± 0.07






EVA/BMA 1X


10
1.26 ± 0.16

 +5%




1X + rapamycin
64
μg
9
0.92 ± 0.14
−27%
−23%




1X + rapamycin
196
μg
10
0.66 ± 0.12* **
−48%
−45%


99020
28 days
Metal


12
1.18 ± 0.10






EVA/BMA 1X + rapamycin
197
μg
8
0.81 ± 0.16

−32%






1Stent nomenclature: EVA/BMA 1X, 2X, and 3X signifies approx. 500 μg, 1000 μg, and 1500 μg total mass (polymer + drug), respectively. TC, top coat of 30 μg, 100 μg, or 300 μg drug-free BMA; Biphasic; 2 × 1X layers of rapamycin in EVA/BMA spearated by a 100 μg drug-free BMA layer.




20.25 mg/kg/d × 14 d preceeded by a loading dose of 0.5 mg/kg/d × 3d prior to stent implantation.



*p < 0.05 from EVA/BMA control. **p < 0.05 from Metal;



#Inflammation score: (0 = essentially no intimal involvement; 1 = <25% intima involved; 2 = ≧25% intima involved; 3 = >50% intima involved).














TABLE 4.0







180 day Porcine Study with Rapamycin-coated stents.


Values are mean ± Standard Error of Mean











Neointimal
% Change
Inflam-



Area
From
mation















Study
Duration
Stent1
Rapamycin
N
(mm2)
Polyme
Metal
Score #


















20007
 3 days
Metal

10
0.38 ± 0.06


1.05 ± 0.06














(ETP-2-002233-P)
1XTC + rapamycin
155 μg
10
0.29 ± 0.03

−24%
1.08 ± 0.04
















 30 days
Metal

9
2.94 ± 0.43


0.11 ± 0.08




1XTC + rapamycin
155 μg
10
1.40 ± 0.11*

−52%*
0.25 ± 0.10



 90 days
Metal

10
3.45 ± 0.34


0.20 ± 0.08




1XTC + rapamycin
155 μg
10
3.03 ± 0.29

−12%
0.80 ± 0.23




1X + rapamycin
171 μg
10
2.86 ± 0.35

−17%
0.60 ± 0.23



180 days
Metal

10
3.65 ± 0.39


0.65 ± 0.21




1XTC + rapamycin
155 μg
10
3.34 ± 0.31

 −8%
1.50 ± 0.34




1X + rapamycin
171 μg
10
3.87 ± 0.28

 +6%
1.68 ± 0.37









The release of rapamycin into the vascular wall of a human from a nonerodible polymeric stent coating provides superior results with respect to the magnitude and duration of the reduction in neointimal hyperplasia within the stent as compared to the vascular walls of animals as set forth above.


Humans implanted with a rapamycin coated stent comprising rapamycin in the same dose range as studied in animal models using the same polymeric matrix, as described above, reveal a much more profound reduction in neointimal hyperplasia than observed in animal models, based on the magnitude and duration of reduction in neointima. The human clinical response to rapamycin reveals essentially total abolition of neointimal hyperplasia inside the stent using both angiographic and intravascular ultrasound measurements. These results are sustained for at least one year as set forth in Table 5 below.









TABLE 5.0







Patients Treated (N = 45 patients) with a Rapamycin-coated Stent










Sirolimus FIM
95%


Effectiveness Measures
(N = 45 Patients, 45 Lesions)
Confidence Limit





Procedure Success (QCA)
100.0% (45/45)
[92.1%, 100.0%]


 4-month In-Stent Diameter Stenosis (%)




Mean ± SD (N)
 4.8% ± 6.1% (30)
[2.6%, 7.0%]


Range (min, max)
(−8.2%, 14.9%)



 6-month In-Stent Diameter Stenosis (%)




Mean ± SD (N)
 8.9% ± 7.6% (13)
[4.8%, 13.0%]


Range (min, max)
(−2.9%, 20.4%)



12-month In-Stent Diameter Stenosis (%)




Mean ± SD (N)
 8.9% ± 6.1% (15)
[5.8%, 12.0%]


Range (min, max)
(−3.0%, 22.0%)



 4-month In-Stent Late Loss (mm)




Mean ± SD (N)
  0.00 ± 0.29 (30)
[−0.10, 0.10]


Range (min, max)
(−0.51, 0.45)



 6-month In-Stent Late Loss (mm)




Mean ± SD (N)
  0.25 ± 0.27 (13)
[0.10, 0.39]


Range (min, max)
(−0.51, 0.91)



12-month In-Stent Late Loss (mm)




Mean ± SD (N)
  0.11 ± 0.36 (15)
[−0.08, 0.29]


Range (min, max)
(−0.51, 0.82)



 4-month Obstruction Volume (%) (IVUS)




Mean ± SD (N)
10.48% ± 2.78% (28)
[9.45%, 11.51%]


Range (min, max)
(4.60%, 16.35%)



 6-month Obstruction Volume (%) (IVUS)




Mean ± SD (N)
 7.22% ± 4.60% (13)
[4.72%, 9.72%],


Range (min, max)
(3.82%, 19.88%)



12-month Obstruction Volume (%) (IVUS)




Mean ± SD (N)
 2.11% ± 5.28% (15)
[0.00%, 4.78%],


Range (min, max)
(0.00%, 19.89%)



 6-month Target Lesion Revascularization (TLR)
 0.0% (0/30)
[0.0%, 9.5%]


12-month Target Lesion Revascularization
 0.0% (0/15)
[0.0%, 18.1%]


(TLR)





QCA = Quantitative Coronary Angiography


SD = Standard Deviation


IVUS = Intravascular Ultrasound






Rapamycin produces an unexpected benefit in humans when delivered from a stent by causing a profound reduction in in-stent neointimal hyperplasia that is sustained for at least one year. The magnitude and duration of this benefit in humans is not predicted from animal model data. Rapamycin used in this context includes rapamycin and all analogs, derivatives and congeners that bind FKBP12 and possess the same pharmacologic properties as rapamycin.


These results may be due to a number of factors. For example, the greater effectiveness of rapamycin in humans is due to greater sensitivity of its mechanism(s) of action toward the pathophysiology of human vascular lesions compared to the pathophysiology of animal models of angioplasty. In addition, the combination of the dose applied to the stent and the polymer coating that controls the release of the drug is important in the effectiveness of the drug.


As stated above, rapamycin reduces vascular hyperplasia by antagonizing smooth muscle proliferation in response to mitogenic signals that are released during angioplasty injury. Also, it is known that rapamycin prevents T-cell proliferation and differentiation when administered systemically. It has also been determined that rapamycin exerts a local inflammatory effect in the vessel wall when administered from a stent in low doses for a sustained period of time (approximately two to six weeks). The local anti-inflammatory benefit is profound and unexpected. In combination with the smooth muscle anti-proliferative effect, this dual mode of action of rapamycin may be responsible for its exceptional efficacy.


Accordingly, rapamycin delivered from a local device platform, reduces neointimal hyperplasia by a combination of anti-inflammatory and smooth muscle anti-proliferative effects. Rapamycin used in this context means rapamycin and all analogs, derivatives and congeners that bind FKBP12 and possess the same pharmacologic properties as rapamycin. Local device platforms include stent coatings, stent sheaths, grafts and local drug infusion catheters or porous balloons or any other suitable means for the in situ or local delivery of drugs, agents or compounds.


The anti-inflammatory effect of rapamycin is evident in data from an experiment, illustrated in Table 6, in which rapamycin delivered from a stent was compared with dexamethasone delivered from a stent. Dexamethasone, a potent steroidal anti-inflammatory agent, was used as a reference standard. Although dexamethasone is able to reduce inflammation scores, rapamycin is far more effective than dexamethasone in reducing inflammation scores. In addition, rapamycin significantly reduces neointimal hyperplasia, unlike dexamethasone.













TABLE 6.0





Group

Neointimal




Rapamycin

Area
% Area
Inflammation


Rap
N =
(mm2)
Stenosis
Score







Uncoated
8
5.24 ± 1.65 
54 ± 19 
0.97 ± 1.00 


Dexamethasone
8
4.31 ± 3.02 
45 ± 31 
0.39 ± 0.24 


(Dex)






Rapamycin
7
2.47 ± 0.94*
26 ± 10*
0.13 ± 0.19*


(Rap)






Rap + Dex
6
2.42 ± 1.58*
26 ± 18*
0.17 ± 0.30*





*= significance level P < 0.05






Rapamycin has also been found to reduce cytokine levels in vascular tissue when delivered from a stent. The data in FIG. 1 illustrates that rapamycin is highly effective in reducing monocyte chemotactic protein (MCP-1) levels in the vascular wall. MCP-1 is an example of a proinflammatory/chemotactic cytokine that is elaborated during vessel injury. Reduction in MCP-1 illustrates the beneficial effect of rapamycin in reducing the expression of proinflammatory mediators and contributing to the anti-inflammatory effect of rapamycin delivered locally from a stent. It is recognized that vascular inflammation in response to injury is a major contributor to the development of neointimal hyperplasia.


Since rapamycin may be shown to inhibit local inflammatory events in the vessel it is believed that this could explain the unexpected superiority of rapamycin in inhibiting neointima.


As set forth above, rapamycin functions on a number of levels to produce such desired effects as the prevention of T-cell proliferation, the inhibition of negative remodeling, the reduction of inflammation, and the prevention of smooth muscle cell proliferation. While the exact mechanisms of these functions are not completely known, the mechanisms that have been identified may be expanded upon.


Studies with rapamycin suggest that the prevention of smooth muscle cell proliferation by blockade of the cell cycle is a valid strategy for reducing neointimal hyperplasia. Dramatic and sustained reductions in late lumen loss and neointimal plaque volume have been observed in patients receiving rapamycin delivered locally from a stent. The present invention expands upon the mechanism of rapamycin to include additional approaches to inhibit the cell cycle and reduce neointimal hyperplasia without producing toxicity.


The cell cycle is a tightly controlled biochemical cascade of events that regulate the process of cell replication. When cells are stimulated by appropriate growth factors, they move from G0 (quiescence) to the G1 phase of the cell cycle. Selective inhibition of the cell cycle in the G1 phase, prior to DNA replication (S phase), may offer therapeutic advantages of cell preservation and viability while retaining anti-proliferative efficacy when compared to therapeutics that act later in the cell cycle i.e. at S, G2 or M phase.


Accordingly, the prevention of intimal hyperplasia in blood vessels and other conduit vessels in the body may be achieved using cell cycle inhibitors that act selectively at the G1 phase of the cell cycle. These inhibitors of the G1 phase of the cell cycle may be small molecules, peptides, proteins, oligonucleotides or DNA sequences. More specifically, these drugs or agents include inhibitors of cyclin dependent kinases (cdk's) involved with the progression of the cell cycle through the G1 phase, in particular cdk2 and cdk4.


Examples of drugs, agents or compounds that act selectively at the G1 phase of the cell cycle include small molecules such as flavopiridol and its structural analogs that have been found to inhibit cell cycle in the late G1 phase by antagonism of cyclin dependent kinases. Therapeutic agents that elevate an endogenous kinase inhibitory proteinkip called P27, sometimes referred to as P27kip1, that selectively inhibits cyclin dependent kinases may be utilized. This includes small molecules, peptides and proteins that either block the degradation of P27 or enhance the cellular production of P27, including gene vectors that can transfact the gene to produce P27. Staurosporin and related small molecules that block the cell cycle by inhibiting protein kinases may be utilized. Protein kinase inhibitors, including the class of tyrphostins that selectively inhibit protein kinases to antagonize signal transduction in smooth muscle in response to a broad range of growth factors such as PDGF and FGF may also be utilized.


Any of the drugs, agents or compounds discussed above may be administered either systemically, for example, orally, intravenously, intramuscularly, subcutaneously, nasally or intradermally, or locally, for example, stent coating, stent covering or local delivery catheter. In addition, the drugs or agents discussed above may be formulated for fast-release or slow release with the objective of maintaining the drugs or agents in contact with target tissues for a period ranging from three days to eight weeks.


As set forth above, the complex of rapamycin and FKPB12 binds to and inhibits a phosphoinositide (PI)-3 kinase called the mammalian Target of Rapamycin or TOR. An antagonist of the catalytic activity of TOR, functioning as either an active site inhibitor or as an allosteric modulator, i.e. an indirect inhibitor that allosterically modulates, would mimic the actions of rapamycin but bypass the requirement for FKBP12. The potential advantages of a direct inhibitor of TOR include better tissue penetration and better physical/chemical stability. In addition, other potential advantages include greater selectivity and specificity of action due to the specificity of an antagonist for one of multiple isoforms of TOR that may exist in different tissues, and a potentially different spectrum of downstream effects leading to greater drug efficacy and/or safety.


The inhibitor may be a small organic molecule (approximate mw<1000), which is either a synthetic or naturally derived product. Wortmanin may be an agent which inhibits the function of this class of proteins. It may also be a peptide or an oligonucleotide sequence. The inhibitor may be administered either sytemically (orally, intravenously, intramuscularly, subcutaneously, nasally, or intradermally) or locally (stent coating, stent covering, local drug delivery catheter). For example, the inhibitor may be released into the vascular wall of a human from a nonerodible polymeric stent coating. In addition, the inhibitor may be formulated for fast-release or slow release with the objective of maintaining the rapamycin or other drug, agent or compound in contact with target tissues for a period ranging from three days to eight weeks.


As stated previously, the implantation of a coronary stent in conjunction with balloon angioplasty is highly effective in treating acute vessel closure and may reduce the risk of restenosis. Intravascular ultrasound studies (Mintz et al., 1996) suggest that coronary stenting effectively prevents vessel constriction and that most of the late luminal loss after stent implantation is due to plaque growth, probably related to neointimal hyperplasia. The late luminal loss after coronary stenting is almost two times higher than that observed after conventional balloon angioplasty. Thus, inasmuch as stents prevent at least a portion of the restenosis process, the use of drugs, agents or compounds which prevent inflammation and proliferation, or prevent proliferation by multiple mechanisms, combined with a stent may provide the most efficacious treatment for post-angioplasty restenosis.


The local delivery of drugs, agents or compounds from a stent has the following advantages; namely, the prevention of vessel recoil and remodeling through the scaffolding action of the stent and the drugs, agents or compounds and the prevention of multiple components of neointimal hyperplasia. This local administration of drugs, agents or compounds to stented coronary arteries may also have additional therapeutic benefit. For example, higher tissue concentrations would be achievable than that which would occur with systemic administration, reduced systemic toxicity, and single treatment and ease of administration. An additional benefit of drug therapy may be to reduce the dose of the therapeutic compounds, thereby limiting their toxicity, while still achieving a reduction in restenosis.


There are a multiplicity of different stents that may be utilized following percutaneous transluminal coronary angioplasty. Although any number of stents may be utilized in accordance with the present invention, for simplicity, one particular stent will be described in exemplary embodiments of the present invention. The skilled artisan will recognize that any number of stents may be utilized in connection with the present invention.


A stent is commonly used as a tubular structure left inside the lumen of a duct to relieve an obstruction. Commonly, stents are inserted into the lumen in a non-expanded form and are then expanded autonomously, or with the aid of a second device in situ. A typical method of expansion occurs through the use of a catheter-mounted angioplasty balloon which is inflated within the stenosed vessel or body passageway in order to shear and disrupt the obstructions associated with the wall components of the vessel and to obtain an enlarged lumen. As set forth below, self-expanding stents may also be utilized.



FIG. 2 illustrates an exemplary stent 100 which may be utilized in accordance with an exemplary embodiment of the present invention. The expandable cylindrical stent 100 comprises a fenestrated structure for placement in a blood vessel, duct or lumen to hold the vessel, duct or lumen open, more particularly for protecting a segment of artery from restenosis after angioplasty. The stent 100 may be expanded circumferentially and maintained in an expanded configuration, that is circumferentially or radially rigid. The stent 100 is axially flexible and when flexed at a band, the stent 100 avoids any externally-protruding component parts.


The stent 100 generally comprises first and second ends with an intermediate section therebetween. The stent 100 has a longitudinal axis and comprises a plurality of longitudinally disposed bands 102, wherein each band 102 defines a generally continuous wave along a line segment parallel to the longitudinal axis. A plurality of circumferentially arranged links 104 maintain the bands 102 in a substantially tubular structure. Essentially, each longitudinally disposed band 102 is connected at a plurality of periodic locations, by a short circumferentially arranged link 104 to an adjacent band 102. The wave associated with each of the bands 102 has approximately the same fundamental spatial frequency in the intermediate section, and the bands 102 are so disposed that the wave associated with them are generally aligned so as to be generally in phase with one another. As illustrated in the figure, each longitudinally arranged band 102 undulates through approximately two cycles before there is a link to an adjacent band.


The stent 100 may be fabricated utilizing any number of methods. For example, the stent 100 may be fabricated from a hollow or formed stainless steel tube that may be machined using lasers, electric discharge milling, chemical etching or other means. The stent 100 is inserted into the body and placed at the desired site in an unexpanded form. In one embodiment, expansion may be effected in a blood vessel by a balloon catheter, where the final diameter of the stent 100 is a function of the diameter of the balloon catheter used.


It should be appreciated that a stent 100 in accordance with the present invention may be embodied in a shape-memory material, including, for example, an appropriate alloy of nickel and titanium. In this embodiment, after the stent 100 has been formed it may be compressed so as to occupy a space sufficiently small as to permit its insertion in a blood vessel or other tissue by insertion means, wherein the insertion means include a suitable catheter, or flexible rod. On emerging from the catheter, the stent 100 may be configured to expand into the desired configuration where the expansion is automatic or triggered by a change in pressure, temperature or electrical stimulation.



FIG. 3 illustrates an exemplary embodiment of the present invention utilizing the stent 100 illustrated in FIG. 2. As illustrated, the stent 100 may be modified to comprise a reservoir 106. Each of the reservoirs may be opened or closed as desired. These reservoirs 106 may be specifically designed to hold the drug, agent, compound or combinations thereof to be delivered. Regardless of the design of the stent 100, it is preferable to have the drug, agent, compound or combinations thereof dosage applied with enough specificity and a sufficient concentration to provide an effective dosage in the lesion area. In this regard, the reservoir size in the bands 102 is preferably sized to adequately apply the drug/drug combination dosage at the desired location and in the desired amount.


In an alternate exemplary embodiment, the entire inner and outer surface of the stent 100 may be coated with various drug and drug combinations in therapeutic dosage amounts. A detailed description of exemplary coating techniques is described below.


Rapamycin or any of the drugs, agents or compounds described above may be incorporated into or affixed to the stent in a number of ways and utilizing any number of biocompatible materials. In the exemplary embodiment, the rapamycin is directly incorporated into a polymeric matrix and sprayed onto the outer surface of the stent. The rapamycin elutes from the polymeric matrix over time and enters the surrounding tissue. The rapamycin preferably remains on the stent for at least three days up to approximately six months and more preferably between seven and thirty days.


Any number of non-erodible polymers may be utilized in conjunction with rapamycin. In the exemplary embodiment, the polymeric matrix comprises two layers. The base layer comprises a solution of ethylene-co-vinylacetate and polybutylmethacrylate. The rapamycin is incorporated into this layer. The outer layer comprises only polybutylmethacrylate and acts as a diffusion barrier to prevent the rapamycin from eluting too quickly and entering the surrounding tissues. The thickness of the outer layer or top coat determines the rate at which the rapamycin elutes from the matrix. Essentially, the rapamycin elutes from the matrix by diffusion through the polymer molecules. Polymers are permeable, thereby allowing solids, liquids and gases to escape therefrom. The total thickness of the polymeric matrix is in the range from about 1 micron to about 20 microns or greater. In a preferred exemplary embodiment, the base layer, including the polymer and drug has a thickness in the range from about 8 microns to about 12 microns and the outer layer has a thickness in the range from about 1 micron to about 2 microns.


The ethylene-co-vinylacetate, polybutylmethacrylate and rapamycin solution may be incorporated into or onto the stent in a number of ways. For example, the solution may be sprayed onto the stent or the stent may be dipped into the solution. In a preferred embodiment, the solution is sprayed onto the stent and then allowed to dry. In another exemplary embodiment, the solution may be electrically charged to one polarity and the stent electrically changed to the opposite polarity. In this manner, the solution and stent will be attracted to one another. In using this type of spraying process, waste may be reduced and more control over the thickness of the coat may be achieved.


Since rapamycin works by entering the surrounding tissue, it is preferably only affixed to the surface of the stent making contact with one tissue. Typically, only the outer surface of the stent makes contact with the tissue. Accordingly, in a preferred embodiment, only the outer surface of the stent is coated with rapamycin. For other drugs, agents or compounds, the entire stent may be coated.


It is important to note that different polymers may be utilized for different stents. For example, in the above-described embodiment, ethylene-co-vinylacetate and polybutylmethacrylate are utilized to form the polymeric matrix. This matrix works well with stainless steel stents. Other polymers may be utilized more effectively with stents formed from other materials, including materials that exhibit superelastic properties such as alloys of nickel and titanium.


Although shown and described is what is believed to be the most practical and preferred embodiments, it is apparent that departures from specific designs and methods described and shown will suggest themselves to those skilled in the art and may be used without departing from the spirit and scope of the invention. The present invention is not restricted to the particular constructions described and illustrated, but should be constructed to cohere with all modifications that may fall within the scope of the appended claims.

Claims
  • 1. A drug delivery device comprising: an intraluminal medical device, the intraluminal medical device including a stent having a fenestrated structure, the stent comprising a plurality of bands and links defining a substantially tubular device with openings;a polymeric coating affixed to the bands and links of the fenestrated structure, the polymeric coating including first and second layers; anda therapeutic dosage of rapamycin incorporated into the polymeric coating for treatment of intimal hyperplasia, constrictive vascular remodeling, and inflammation caused by injury, the rapamycin being incorporated into the first layer of the polymeric coating, the first layer comprising a solution of ethylene-co-vinylacetate and polybutylmethacrylate, and the second layer, comprising polybutylmethacrylate and acting as a diffusion layer, the first layer having a thickness in the range from about eight microns to about twelve microns and the second layer having a thickness in the range from about one micron to about two microns, the rapamycin being configured for release over a period of about seven to thirty days and having a dosage in the range from about 35 to 430 micrograms per 15 to 18 mm length stents thereby producing a peak 50 to 55 percent reduction in neointimal hyperplasia.
CROSS REFERENCE TO RELATED APPLICATIONS

This application is a continuation-in-part of U.S. application Ser. No. 09/850,365 filed on May 7, 2001 now abandoned, which is a continuation-in-part application of U.S. application Ser. No. 09/575,480, filed on May 19, 2000, U.S. Pat. No. 8,029,561 which claims the benefit of U.S. Provisional Application No. 60/204,417, filed May 12, 2000 and claims the benefit of U.S. Provisional Application No. 60/262,614, filed Jan. 18, 2001, U.S. Provisional Application No. 60/262,461, filed Jan. 18, 2001, U.S. Provisional Application No. 60/263,806, filed Jan. 24, 2001 and U.S. Provisional Application No. 60/263,979, filed Jan. 25, 2001.

US Referenced Citations (519)
Number Name Date Kind
861659 Johnston Jul 1907 A
3051677 Rexford Aug 1962 A
3279996 Long et al. Oct 1966 A
3526005 Bokros Sep 1970 A
3599641 Sheridan Aug 1971 A
3657744 Ersek Apr 1972 A
3744596 Sander Jul 1973 A
3779805 Alsberg Dec 1973 A
3929992 Sehgal et al. Dec 1975 A
3932627 Margraf Jan 1976 A
3948254 Zaffaroni Apr 1976 A
3952334 Bokros et al. Apr 1976 A
3968800 Vilasi Jul 1976 A
4069307 Higuchi et al. Jan 1978 A
4076285 Martinez Feb 1978 A
4292965 Nash et al. Oct 1981 A
4299226 Banka Nov 1981 A
4300244 Bokros Nov 1981 A
4312920 Pierce et al. Jan 1982 A
4321711 Mano Mar 1982 A
4323071 Simpson et al. Apr 1982 A
4390599 Broyles Jun 1983 A
4413359 Akiyama et al. Nov 1983 A
4423183 Close Dec 1983 A
4441216 Ionescu et al. Apr 1984 A
4503569 Dotter Mar 1985 A
4512338 Balko et al. Apr 1985 A
4550447 Seiler, Jr. et al. Nov 1985 A
4553545 Maass et al. Nov 1985 A
4560374 Hammerslag Dec 1985 A
4562596 Kronberg Jan 1986 A
4565740 Golander et al. Jan 1986 A
4580568 Gianturco Apr 1986 A
4613665 Larm Sep 1986 A
4642111 Sakamoto et al. Feb 1987 A
4655771 Wallsten Apr 1987 A
4656083 Hoffman et al. Apr 1987 A
4676241 Webb et al. Jun 1987 A
4678466 Rosenwald Jul 1987 A
4687482 Hanson Aug 1987 A
4689046 Bokros Aug 1987 A
4731054 Billeter et al. Mar 1988 A
4733665 Palmaz Mar 1988 A
4739762 Palmaz Apr 1988 A
4740207 Kreamer Apr 1988 A
4749585 Greco et al. Jun 1988 A
4753652 Langer et al. Jun 1988 A
4760849 Kropf Aug 1988 A
4768507 Fischell et al. Sep 1988 A
4776337 Palmaz Oct 1988 A
4786500 Wong Nov 1988 A
4787899 Lazarus Nov 1988 A
4800882 Gianturco Jan 1989 A
4810784 Larm Mar 1989 A
4856516 Hillstead Aug 1989 A
4871357 Hsu et al. Oct 1989 A
4872867 Joh Oct 1989 A
4876109 Mayer et al. Oct 1989 A
4886062 Wiktor Dec 1989 A
4907336 Gianturco Mar 1990 A
4916193 Tang et al. Apr 1990 A
4954126 Wallsten Sep 1990 A
4961954 Goldberg et al. Oct 1990 A
4969458 Wiktor Nov 1990 A
4990131 Dardik Feb 1991 A
4990155 Wilkoff Feb 1991 A
4994071 MacGregor Feb 1991 A
4994298 Yasuda Feb 1991 A
4998923 Samson et al. Mar 1991 A
5015253 MacGregor May 1991 A
5019090 Pinchuk May 1991 A
5019096 Fox, Jr. et al. May 1991 A
5029877 Fedeli et al. Jul 1991 A
5034265 Hoffman et al. Jul 1991 A
5035706 Gianturco Jul 1991 A
5041100 Rowland et al. Aug 1991 A
5041126 Gianturco Aug 1991 A
5047020 Hsu Sep 1991 A
5049132 Shaffer et al. Sep 1991 A
5049403 Larm et al. Sep 1991 A
5053048 Pinchuk Oct 1991 A
5059166 Fischell et al. Oct 1991 A
5061275 Wallsten et al. Oct 1991 A
5061750 Feijen et al. Oct 1991 A
5064435 Porter Nov 1991 A
5092877 Pinchuk Mar 1992 A
5102417 Palmaz Apr 1992 A
5104404 Wolff Apr 1992 A
5116365 Hillstead May 1992 A
5122154 Rhodes Jun 1992 A
5131908 Dardik et al. Jul 1992 A
5133732 Wiktor Jul 1992 A
5134192 Feijen et al. Jul 1992 A
5135536 Hillstead Aug 1992 A
5163952 Froix Nov 1992 A
5163958 Pinchuk Nov 1992 A
5171217 March et al. Dec 1992 A
5171262 MacGregor Dec 1992 A
5176660 Truckai Jan 1993 A
5176972 Bloom et al. Jan 1993 A
5178618 Kandarpa Jan 1993 A
5180366 Woods Jan 1993 A
5182317 Winters et al. Jan 1993 A
5185408 Tang et al. Feb 1993 A
5192307 Wall Mar 1993 A
5195984 Schatz Mar 1993 A
5199951 Spears Apr 1993 A
5202332 Hughes et al. Apr 1993 A
5213576 Abiuso et al. May 1993 A
5213898 Larm et al. May 1993 A
5217483 Tower Jun 1993 A
5222971 Willard et al. Jun 1993 A
5226913 Pinchuk Jul 1993 A
5234456 Silvestrini Aug 1993 A
5246445 Yachia et al. Sep 1993 A
5252579 Skotnicki et al. Oct 1993 A
5256790 Nelson Oct 1993 A
5258020 Froix Nov 1993 A
5258021 Duran Nov 1993 A
5262451 Winters et al. Nov 1993 A
5266073 Wall Nov 1993 A
5272012 Opolski Dec 1993 A
5275622 Lazarus et al. Jan 1994 A
5282823 Schwartz et al. Feb 1994 A
5282824 Gianturco Feb 1994 A
5283257 Gregory et al. Feb 1994 A
5288711 Mitchell et al. Feb 1994 A
5290305 Inoue Mar 1994 A
5292331 Boneau Mar 1994 A
5292802 Rhee et al. Mar 1994 A
5304121 Sahatjian Apr 1994 A
5304200 Spaulding Apr 1994 A
5306250 March et al. Apr 1994 A
5308862 Ohlstein May 1994 A
5308889 Rhee et al. May 1994 A
5314444 Gianturco May 1994 A
5314472 Fontaine May 1994 A
5328471 Slepian Jul 1994 A
5334301 Heinke et al. Aug 1994 A
5336518 Narayanan et al. Aug 1994 A
5338770 Winters et al. Aug 1994 A
5342348 Kaplan Aug 1994 A
5342387 Summers Aug 1994 A
5342621 Eury Aug 1994 A
5354257 Roubin et al. Oct 1994 A
5354308 Simon et al. Oct 1994 A
5356433 Rowland et al. Oct 1994 A
5362718 Skotnicki et al. Nov 1994 A
5366504 Andersen et al. Nov 1994 A
5368566 Crocker Nov 1994 A
5370683 Fontaine Dec 1994 A
5370691 Samson Dec 1994 A
5375612 Cottenceau et al. Dec 1994 A
5376112 Duran Dec 1994 A
5378475 Smith et al. Jan 1995 A
5378836 Kao et al. Jan 1995 A
5380299 Fearnot et al. Jan 1995 A
5382261 Palmaz Jan 1995 A
5383853 Jung et al. Jan 1995 A
5383928 Scott et al. Jan 1995 A
5385908 Nelson et al. Jan 1995 A
5385909 Nelson et al. Jan 1995 A
5385910 Ocain et al. Jan 1995 A
5387235 Chuter Feb 1995 A
5387680 Nelson Feb 1995 A
5389106 Tower Feb 1995 A
5389639 Failli et al. Feb 1995 A
5391730 Skotnicki et al. Feb 1995 A
5393772 Yue et al. Feb 1995 A
5395390 Simon et al. Mar 1995 A
5397355 Marin et al. Mar 1995 A
5399352 Hanson Mar 1995 A
5403341 Solar Apr 1995 A
5405377 Cragg Apr 1995 A
5409696 Narayanan et al. Apr 1995 A
5411549 Peters May 1995 A
5415619 Lee et al. May 1995 A
5417969 Hsu et al. May 1995 A
5419760 Narciso, Jr. May 1995 A
D359802 Fontaine Jun 1995 S
5421955 Lau Jun 1995 A
5423885 Williams Jun 1995 A
5429618 Keogh Jul 1995 A
5429634 Narcisco Jul 1995 A
5439446 Barry Aug 1995 A
5441515 Khosravi et al. Aug 1995 A
5441516 Wang et al. Aug 1995 A
5441947 Dodge et al. Aug 1995 A
5441977 Russo et al. Aug 1995 A
5443458 Eury Aug 1995 A
5443477 Marin et al. Aug 1995 A
5443496 Schwartz et al. Aug 1995 A
5443498 Fontaine Aug 1995 A
5443500 Sigwart Aug 1995 A
5447724 Helmus et al. Sep 1995 A
5449372 Schmaltz et al. Sep 1995 A
5449373 Pinchasik et al. Sep 1995 A
5449382 Dayton Sep 1995 A
5464450 Buscemi et al. Nov 1995 A
5464540 Friesen et al. Nov 1995 A
5464650 Berg et al. Nov 1995 A
5472985 Grainger et al. Dec 1995 A
5474563 Myler et al. Dec 1995 A
5486357 Narayanan Jan 1996 A
5491231 Nelson et al. Feb 1996 A
5496365 Sgro Mar 1996 A
5500013 Buscemi et al. Mar 1996 A
5504091 Molnar-Kimber et al. Apr 1996 A
5508286 Skotnicki et al. Apr 1996 A
5510077 Dinh et al. Apr 1996 A
5512055 Domb et al. Apr 1996 A
5514680 Weber et al. May 1996 A
5516781 Morris et al. May 1996 A
5519042 Morris et al. May 1996 A
5523092 Hanson et al. Jun 1996 A
5527354 Fontaine et al. Jun 1996 A
5541191 Skotnicki et al. Jul 1996 A
5545208 Wolff et al. Aug 1996 A
5551954 Buscemi et al. Sep 1996 A
5554182 Dinh et al. Sep 1996 A
5554954 Takahashi Sep 1996 A
5556413 Lam Sep 1996 A
5559122 Nelson et al. Sep 1996 A
5562922 Lambert Oct 1996 A
5563145 Failli et al. Oct 1996 A
5563146 Morris et al. Oct 1996 A
5569197 Helmus et al. Oct 1996 A
5569295 Lam Oct 1996 A
5569462 Martinson et al. Oct 1996 A
5569463 Helmus et al. Oct 1996 A
5571089 Crocker Nov 1996 A
5571166 Dinh et al. Nov 1996 A
5574059 Regunathan et al. Nov 1996 A
5575818 Pinchuk Nov 1996 A
5578075 Dayton Nov 1996 A
5580873 Bianco et al. Dec 1996 A
5580874 Bianco et al. Dec 1996 A
5591140 Narayanan et al. Jan 1997 A
5591197 Orth et al. Jan 1997 A
5591224 Schwartz et al. Jan 1997 A
5591227 Dinh et al. Jan 1997 A
5599352 Dinh et al. Feb 1997 A
5599844 Grainger et al. Feb 1997 A
5603722 Phan et al. Feb 1997 A
5605696 Eury et al. Feb 1997 A
5607463 Schwartz et al. Mar 1997 A
5607475 Cahalan et al. Mar 1997 A
5609629 Fearnot et al. Mar 1997 A
5616608 Kinsella et al. Apr 1997 A
5618837 Hart et al. Apr 1997 A
5620984 Bianco et al. Apr 1997 A
5621102 Bianco et al. Apr 1997 A
5622975 Singh et al. Apr 1997 A
5624411 Tuch Apr 1997 A
5628785 Schwartz et al. May 1997 A
5629077 Turnlund et al. May 1997 A
5629315 Bianco et al. May 1997 A
5632763 Glastra May 1997 A
5632771 Boatman et al. May 1997 A
5632776 Kurumatani et al. May 1997 A
5632840 Campbell May 1997 A
5635201 Fabo Jun 1997 A
5637113 Tartaglia et al. Jun 1997 A
5643312 Fischell et al. Jul 1997 A
5643939 Ohlstein Jul 1997 A
5646160 Morris et al. Jul 1997 A
5648357 Bianco et al. Jul 1997 A
5649952 Lam Jul 1997 A
5649977 Campbell Jul 1997 A
5651174 Schwartz et al. Jul 1997 A
5652243 Bianco et al. Jul 1997 A
5653747 Dereume Aug 1997 A
5653992 Bezwada et al. Aug 1997 A
5660873 Nikolaychik et al. Aug 1997 A
5662609 Slepian Sep 1997 A
5665591 Sonenshein et al. Sep 1997 A
5665728 Morris et al. Sep 1997 A
5665772 Cottens et al. Sep 1997 A
5667764 Kopia et al. Sep 1997 A
5669924 Shaknovich Sep 1997 A
5670506 Leigh et al. Sep 1997 A
5674242 Phan et al. Oct 1997 A
5679400 Tuch Oct 1997 A
5679659 Verhoeven et al. Oct 1997 A
5684061 Ohnishi et al. Nov 1997 A
5691311 Marganore et al. Nov 1997 A
5693085 Buirge et al. Dec 1997 A
5697967 Dinh et al. Dec 1997 A
5697971 Fischell et al. Dec 1997 A
5700286 Tartaglia et al. Dec 1997 A
5707385 Williams Jan 1998 A
5709874 Hanson et al. Jan 1998 A
5710174 West et al. Jan 1998 A
5713949 Jayaraman Feb 1998 A
5716981 Hunter et al. Feb 1998 A
5725549 Lam Mar 1998 A
5725567 Wolff et al. Mar 1998 A
5728150 McDonald et al. Mar 1998 A
5728420 Keogh Mar 1998 A
5731326 Hart et al. Mar 1998 A
5733327 Igaki et al. Mar 1998 A
5733920 Mansuri et al. Mar 1998 A
5733925 Kunz et al. Mar 1998 A
5735897 Buirge Apr 1998 A
5739138 Bianco et al. Apr 1998 A
5744587 Alaska et al. Apr 1998 A
5755734 Richter et al. May 1998 A
5755772 Evans et al. May 1998 A
5759205 Valentini Jun 1998 A
5769883 Buscemi et al. Jun 1998 A
5776184 Tuch Jul 1998 A
5780462 Lee et al. Jul 1998 A
5780476 Underiner et al. Jul 1998 A
5782908 Cahalan et al. Jul 1998 A
5786171 Lee et al. Jul 1998 A
5788979 Alt Aug 1998 A
5792106 Mische Aug 1998 A
5792772 Bianco et al. Aug 1998 A
5798372 Davies et al. Aug 1998 A
5799384 Schwartz et al. Sep 1998 A
5800507 Schwartz Sep 1998 A
5800508 Goicoechea et al. Sep 1998 A
5807743 Stinchcomb et al. Sep 1998 A
5807861 Klein et al. Sep 1998 A
5811447 Kunz et al. Sep 1998 A
5820917 Tuch Oct 1998 A
5820918 Ronan et al. Oct 1998 A
5824045 Alt Oct 1998 A
5824048 Tuch Oct 1998 A
5824049 Ragheb et al. Oct 1998 A
5827587 Fukushi Oct 1998 A
5827734 Weigle et al. Oct 1998 A
5833651 Donovan et al. Nov 1998 A
5837008 Berg et al. Nov 1998 A
5837313 Ding et al. Nov 1998 A
5840009 Fischell et al. Nov 1998 A
5843120 Israel et al. Dec 1998 A
5843166 Lentz et al. Dec 1998 A
5843172 Yan Dec 1998 A
5849034 Schwartz Dec 1998 A
5851217 Wolff et al. Dec 1998 A
5851231 Wolff et al. Dec 1998 A
5858967 Weigle et al. Jan 1999 A
5858990 Walsh Jan 1999 A
5861027 Trapp Jan 1999 A
5865814 Tuch Feb 1999 A
5871535 Wolff et al. Feb 1999 A
5873904 Ragheb et al. Feb 1999 A
5876433 Lunn Mar 1999 A
5877224 Brocchini et al. Mar 1999 A
5879697 Ding et al. Mar 1999 A
5882335 Leone et al. Mar 1999 A
5883110 Tang et al. Mar 1999 A
5891108 Leone et al. Apr 1999 A
5893840 Hull et al. Apr 1999 A
5897911 Loeffler Apr 1999 A
5900246 Lambert May 1999 A
5902266 Leone et al. May 1999 A
5912253 Cottens et al. Jun 1999 A
5916910 Lai Jun 1999 A
5922393 Jayaraman Jul 1999 A
5922730 Hu et al. Jul 1999 A
5932243 Fricker et al. Aug 1999 A
5932299 Katoot Aug 1999 A
5932580 Levitzki et al. Aug 1999 A
5951586 Berg et al. Sep 1999 A
5957971 Schwartz Sep 1999 A
5958949 Hamilton et al. Sep 1999 A
5959075 Lok et al. Sep 1999 A
5962265 Norris et al. Oct 1999 A
5968091 Pinchuk et al. Oct 1999 A
5972027 Johnson Oct 1999 A
5976534 Hart et al. Nov 1999 A
5977163 Li et al. Nov 1999 A
5980553 Gray et al. Nov 1999 A
5980566 Alt et al. Nov 1999 A
5980972 Ding Nov 1999 A
5981568 Kunz et al. Nov 1999 A
5985307 Hanson et al. Nov 1999 A
5986049 Forstrom et al. Nov 1999 A
5997468 Wolff et al. Dec 1999 A
6004346 Wolff et al. Dec 1999 A
6015432 Rakos et al. Jan 2000 A
6015815 Mollison Jan 2000 A
6039721 Johnson et al. Mar 2000 A
6059813 Vrba et al. May 2000 A
6071305 Brown et al. Jun 2000 A
6074659 Kunz et al. Jun 2000 A
6080190 Schwartz Jun 2000 A
6096070 Ragheb et al. Aug 2000 A
6120536 Dinge et al. Sep 2000 A
6120847 Yang et al. Sep 2000 A
6136798 Cody et al. Oct 2000 A
6140127 Sprague Oct 2000 A
6146358 Rowe Nov 2000 A
6153252 Hossainy et al. Nov 2000 A
6159488 Nagier et al. Dec 2000 A
6171232 Papandreou et al. Jan 2001 B1
6171609 Kunz Jan 2001 B1
6177272 Nabel et al. Jan 2001 B1
6179817 Zhong Jan 2001 B1
6187757 Clackson et al. Feb 2001 B1
6193746 Strecker Feb 2001 B1
6214901 Chudzik et al. Apr 2001 B1
6225346 Tang et al. May 2001 B1
6240616 Yan Jun 2001 B1
6245537 Williams et al. Jun 2001 B1
6251920 Grainger et al. Jun 2001 B1
6254632 Wu et al. Jul 2001 B1
6254634 Anderson et al. Jul 2001 B1
6258121 Yang et al. Jul 2001 B1
6268390 Kunz Jul 2001 B1
6273913 Wright et al. Aug 2001 B1
6284305 Ding et al. Sep 2001 B1
6287320 Slepian Sep 2001 B1
6287628 Hossainy et al. Sep 2001 B1
6299604 Ragheb et al. Oct 2001 B1
6306144 Sydney et al. Oct 2001 B1
6306166 Barry et al. Oct 2001 B1
6306176 Whitbourne Oct 2001 B1
6306421 Kunz et al. Oct 2001 B1
6309380 Larson et al. Oct 2001 B1
6309660 Hsu et al. Oct 2001 B1
6313264 Caggiano et al. Nov 2001 B1
6316018 Ding et al. Nov 2001 B1
6335029 Kamath et al. Jan 2002 B1
6358556 Ding et al. Mar 2002 B1
6368658 Schwartz et al. Apr 2002 B1
6369039 Palasis et al. Apr 2002 B1
6379382 Yang Apr 2002 B1
6384046 Schuler et al. May 2002 B1
6387121 Alt May 2002 B1
6403635 Kinsella et al. Jun 2002 B1
6407067 Schafer Jun 2002 B1
6448221 Sheppard et al. Sep 2002 B1
6471979 New et al. Oct 2002 B2
6492106 Sabatini et al. Dec 2002 B1
6517858 Le Moel et al. Feb 2003 B1
6517889 Jayaraman Feb 2003 B1
6545097 Pinchuk et al. Apr 2003 B2
6585764 Wright et al. Jul 2003 B2
6620194 Ding et al. Sep 2003 B2
6623521 Steinke et al. Sep 2003 B2
6627246 Mehta et al. Sep 2003 B2
6673385 Ding et al. Jan 2004 B1
6742570 Kono Jun 2004 B2
6746773 Llanos et al. Jun 2004 B2
6753071 Pacetti Jun 2004 B1
6776796 Falotico et al. Aug 2004 B2
6808536 Wright et al. Oct 2004 B2
6818247 Chen et al. Nov 2004 B1
6863685 Davila et al. Mar 2005 B2
6872225 Rowan et al. Mar 2005 B1
6884429 Koziak et al. Apr 2005 B2
6939375 Sirhan et al. Sep 2005 B2
6949514 Wallner et al. Sep 2005 B2
7018405 Sirhan et al. Mar 2006 B2
7048939 Elkins et al. May 2006 B2
7056339 Elkins et al. Jun 2006 B2
7056550 Davila et al. Jun 2006 B2
7078240 Li et al. Jul 2006 B2
7087078 Hildebrand et al. Aug 2006 B2
7135039 De Scheerder et al. Nov 2006 B2
7195640 Falotico et al. Mar 2007 B2
7217286 Falotico et al. May 2007 B2
7223286 Wright et al. May 2007 B2
7229473 Falotico et al. Jun 2007 B2
7261735 Llanos et al. Aug 2007 B2
7300662 Falotico et al. Nov 2007 B2
7419678 Falotico Sep 2008 B2
20010007083 Roorda Jul 2001 A1
20010029351 Falotico et al. Oct 2001 A1
20010029660 Johnson Oct 2001 A1
20010032014 Yang et al. Oct 2001 A1
20010034363 Li et al. Oct 2001 A1
20010037145 Guruwaiya et al. Nov 2001 A1
20020007215 Falotico Jan 2002 A1
20020010418 Lary et al. Jan 2002 A1
20020032477 Helmus et al. Mar 2002 A1
20020041899 Chudzik et al. Apr 2002 A1
20020061326 Li et al. May 2002 A1
20020068969 Shanley et al. Jun 2002 A1
20020071902 Ding et al. Jun 2002 A1
20020082680 Shanley et al. Jun 2002 A1
20020082685 Sirhan et al. Jun 2002 A1
20020091433 Ding et al. Jul 2002 A1
20020095114 Palasis Jul 2002 A1
20020099438 Furst Jul 2002 A1
20020103505 Thompson Aug 2002 A1
20020103526 Steinke Aug 2002 A1
20020119178 Levesque et al. Aug 2002 A1
20020123505 Mollison et al. Sep 2002 A1
20020127327 Schwarz et al. Sep 2002 A1
20020133222 Das Sep 2002 A1
20020133224 Bajgar et al. Sep 2002 A1
20020165608 Llanos Nov 2002 A1
20020193475 Hossainy et al. Dec 2002 A1
20030065377 Davila et al. Apr 2003 A1
20030099712 Jayaraman May 2003 A1
20030216699 Falotico Nov 2003 A1
20040049265 Ding et al. Mar 2004 A1
20040243097 Falotico et al. Dec 2004 A1
20040260268 Falotico et al. Dec 2004 A1
20050002986 Falotico et al. Jan 2005 A1
20050004663 Llanos et al. Jan 2005 A1
20050033261 Falotico et al. Feb 2005 A1
20050106210 Ding et al. May 2005 A1
20050136090 Falotico et al. Jun 2005 A1
20050158360 Falotico et al. Jul 2005 A1
20050187611 Ding et al. Aug 2005 A1
20050208200 Ding et al. Sep 2005 A1
20060088654 Ding et al. Apr 2006 A1
20060089705 Ding et al. Apr 2006 A1
20060222756 Davila et al. Oct 2006 A1
20070026036 Falotico et al. Feb 2007 A1
20070087028 Falotico et al. Apr 2007 A1
20070276474 Llanos et al. Nov 2007 A1
20070276475 Llanos et al. Nov 2007 A1
20070276476 Llanos et al. Nov 2007 A1
Foreign Referenced Citations (79)
Number Date Country
3205942 Sep 1983 DE
19723723 Dec 1998 DE
0 145 166 Jun 1985 EP
0 177 330 Apr 1986 EP
0 183 372 Jun 1986 EP
0 221 570 May 1987 EP
0 421 729 Apr 1991 EP
0 540 290 Oct 1992 EP
0 568 310 Nov 1993 EP
0 604 022 Jun 1994 EP
0 621 015 Oct 1994 EP
0 623 354 Nov 1994 EP
0 734 698 Mar 1996 EP
0 712 615 May 1996 EP
0 716 836 Jun 1996 EP
0 800 801 Aug 1996 EP
0 731 721 Oct 1996 EP
0 747 069 Dec 1996 EP
0 761 251 Mar 1997 EP
0 830 853 Jul 1997 EP
0 815 803 Jul 1998 EP
0 850 651 Jul 1998 EP
0 938 878 Sep 1999 EP
0950386 Oct 1999 EP
0950386 Oct 1999 EP
0 968 688 Jan 2000 EP
0 633 032 Feb 2001 EP
1 192 957 Apr 2002 EP
1 588 726 Oct 2005 EP
1 588 727 Oct 2005 EP
0 566 807 Apr 1992 FR
1 205 743 Sep 1970 GB
2 135 585 Sep 1984 GB
0 662 307 Dec 1994 GB
660689 May 1979 SU
WO 8903232 Apr 1989 WO
WO 9112779 Sep 1991 WO
WO 9117724 Nov 1991 WO
WO 9215286 Sep 1992 WO
WO 9401056 Jan 1994 WO
WO 9421308 Sep 1994 WO
WO 9421309 Sep 1994 WO
WO 9424961 Nov 1994 WO
WO 9600272 Jan 1996 WO
WO 9626689 Sep 1996 WO
WO 9632907 Oct 1996 WO
WO 9634580 Nov 1996 WO
WO 9641807 Dec 1996 WO
WO 9725000 Jul 1997 WO
WO 9733534 Sep 1997 WO
WO 9735575 Oct 1997 WO
WO 9808463 Mar 1998 WO
WO 9813344 Apr 1998 WO
WO 9819628 May 1998 WO
WO 9823228 Jun 1998 WO
WO 9823244 Jun 1998 WO
WO 9834669 Aug 1998 WO
WO 9836784 Aug 1998 WO
WO 9847447 Oct 1998 WO
WO 9856312 Dec 1998 WO
WO 9955396 Nov 1999 WO
WO 0021584 Apr 2000 WO
0027445 May 2000 WO
WO 0027445 May 2000 WO
WO 0032255 Jun 2000 WO
WO 0038754 Jul 2000 WO
WO 0187342 Nov 2001 WO
WO 0187372 Nov 2001 WO
WO 0187373 Nov 2001 WO
WO 0187376 Nov 2001 WO
WO 0226139 Apr 2002 WO
WO 0226271 Apr 2002 WO
WO 0226280 Apr 2002 WO
WO 0226281 Apr 2002 WO
WO 02087472 Nov 2002 WO
WO 03015664 Apr 2003 WO
WO 03057218 Jul 2003 WO
WO 03082368 Oct 2003 WO
WO 2004011465 Feb 2004 WO
Related Publications (1)
Number Date Country
20050033261 A1 Feb 2005 US
Provisional Applications (5)
Number Date Country
60204417 May 2000 US
60262614 Jan 2001 US
60262461 Jan 2001 US
60263806 Jan 2001 US
60263979 Jan 2001 US
Continuation in Parts (2)
Number Date Country
Parent 09850365 May 2001 US
Child 10833200 US
Parent 09575480 May 2000 US
Child 09850365 US