25.10.2014 Views

Selecta® 7000 - Lumenis Ophthalmology

Selecta® 7000 - Lumenis Ophthalmology

Selecta® 7000 - Lumenis Ophthalmology

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

®<br />

®<br />

Selecta® <strong>7000</strong><br />

FDA PREMARKET NOTIFICATION 510(K)<br />

CLINICAL PERFORMANCE REPORT


1.0 | Background and Rationale for SLT<br />

Lasers have been employed in the management of glaucoma for over two decades, in a range of<br />

procedures that include iridotomy, gonioplasty, goniotomy, sclerostomy, trabecular ablation and<br />

trabeculoplasty. The early work of Worthen and Wickham 1,2 established that a continuous wave argon<br />

laser could be safely used to photocoagulate the trabecular meshwork, with significant decreases in<br />

intraocular pressure that were maintained over a period of months. Several years later, Wise and<br />

Witter, 3 using lower laser energy in the procedure known as argon laser trabeculoplasty or ALT,<br />

reported successful reduction of intraocular pressure in a population of patients who had failed<br />

medical therapy.<br />

ALT was initially adopted as an alternative or adjunct to medical therapy in order to reduce intraocular<br />

pressure in patients who had failed medical therapy and thereby postpone or obviate the need for<br />

surgery. Over time, as the pressure-lowering effect of and safety of ALT were confirmed, use of this<br />

procedure as the starting point for management of primary open angle glaucoma emerged. In the<br />

Glaucoma Laser Trial, 4-7 initial treatment with argon laser trabeculoplasty was shown to have a greater<br />

pressure-lowering effect than medication (timolol maleate 0.5%) at 2 years of follow-up. Additionally,<br />

at two years of follow-up, fewer of the laser-treated eyes required simultaneous administration of two<br />

or more glaucoma medications to control intraocular pressure. 5 While there were no major differences<br />

between the two treatment arms with respect to changes in visual acuity or visual field over the first<br />

2 years of follow-up, at 3.5 years the mean threshold per test location of the visual field was higher for<br />

eyes initially treated with ALT than for the eyes initially treated with medication. 6 The results at 5<br />

years of follow-up were consistent with the earlier outcome, with respect to intraocular pressure level,<br />

visual field results, optic disc status, visual acuity and medication use, which continued to be lower in<br />

the eyes initially treated with laser. While none of the differences between the treatment groups were<br />

large, all of these differences suggested that initial treatment with ALT in patients with primary openangle<br />

glaucoma was at least as effective as intervention with timolol. 7<br />

Although ALT has been shown to be a viable alternative to initial treatment with medications, its use<br />

in this population has been limited by several factors, particularly the underlying coagulative damage<br />

to the trabecular meshwork induced by the procedure. It has been established that laser trabeculoplasty<br />

reduces intraocular pressure by increasing aqueous outflow, 8-10 however the mechanism for<br />

improved outflow has not been fully elucidated. Two general hypotheses have been advanced to<br />

explain the increase in aqueous outflow following laser trabeculoplasty. One of these suggested<br />

mechanisms is that ALT induces a biologic response in the trabecular meshwork which involves<br />

increased trabecular cell division and migration of trabecular cells to the burn areas, and phagocytosis<br />

of debris, production of glycosaminoglycans (GAGs), and activation of metalloproteases involved in<br />

the maintenance of the extracellular matrix, all of which are normal functions of the trabecular<br />

meshwork cells. 11,12 This has been supported by Alvarado and Murphy, 13 who examined outflow<br />

obstruction in pigmentary and primary open angle glaucoma and demonstrated that macrophages<br />

are the major cell type responsible for trabecular meshwork clearance of pigment and debris.<br />

1


21.0 | Background and Rationale for SL T<br />

While the role of the biologic response elicited by ALT continues to be a subject of investigation,<br />

the mechanical and thermal effects on the trabecular meshwork tissue following ALT have been well<br />

documented. Histopathologic and scanning electron microscopic studies of the trabecular meshwork<br />

suggest that laser energy may induce an inner bowing of the trabecular meshwork, adequate to open<br />

Schlemm’s canal. 14 However, this has not been confirmed by other investigators in histologic analysis<br />

of laser-treated cadaver eyes. 15 Furthermore, while some of the effects observed histologically may<br />

increase aqueous outflow, others may be responsible for the reported failures of the ALT procedure.<br />

Coagulative necrosis caused by thermal injury of the treated tissue, and disruption of trabecular<br />

beams with fragmented cells and fibrocellular tissue in the juxtacanalicular region of the trabecular<br />

meshwork were observed in cynomolgus monkeys following ALT. 16 In ALT-treated human eyes,<br />

histopathologic examination of samples obtained at trabeculectomy revealed early changes similar to<br />

those observed in monkeys, with disruption of trabecular beams, and accumulation of cellular and<br />

fibrinous debris. 14 In tissue samples obtained 6 to 12 months following ALT, confluent areas of fibrosis<br />

and occlusion of the trabecular spaces by abnormally migrating corneal endothelial cells were<br />

observed. 14 Such changes in the architecture of the trabecular meshwork ultimately result in trabecular<br />

fusion and/or occlusion of the trabecular spaces, leading to obstruction of aqueous outflow, as<br />

demonstrated by Melamed 16 in studies in primates which established that aqueous flow is reduced in<br />

the area treated with argon laser trabeculoplasty. These changes may cause or contribute to the elevated<br />

post-treatment intraocular pressure observed following ALT, and to the limited success of ALT retreatment.<br />

17-21 In studies evaluating the outcome of ALT retreatment, only a very limited proportion of<br />

patients experienced a reduction of intraocular pressure. Repeat ALT appears to have little chance of<br />

success in eyes that previously failed to respond to treatment for a prolonged period of time, and may<br />

be followed by such a substantial increase in intraocular pressure that immediate surgical intervention<br />

is required. 17,19<br />

Thus, the ultrastructural changes of the human trabecular meshwork following ALT comprise both<br />

stimulating and destructive events. While laser-stimulated trabecular cell division after ALT may have<br />

a positive effect on outflow resistance by causing alterations in the synthesis of the proteoglycan<br />

components of the extracellular matrix, 22 the destructive mechanisms and excessive repair response<br />

associated with ALT are related to treatment failure. These limitations of ALT, related primarily to<br />

its coagulative effect on the trabecular meshwork, have led to interest in the development of laser<br />

procedures that may be less destructive to the tissue of the trabecular meshwork while retaining the<br />

therapeutic benefits of laser irradiation. In the ALT procedure, performed with continuous wave<br />

lasers with pulse durations in the millisecond range or greater, although laser energy is absorbed<br />

primarily by pigment-containing cells of the trabecular meshwork, there is thermal dissipation from<br />

the pigmented cells to surrounding tissues, causing thermal coagulation of tissue. This thermally<br />

mediated damage to contiguous tissues can be limited by means of selective photothermolysis, which<br />

relies on selective absorption of a short laser pulse to generate and confine heat to pigmented targets.


These targets must have greater optical absorption than the surrounding tissues. Initially developed<br />

with oxyhemoglobin as the target pigment, selective targeting or photothermolysis has been utilized<br />

in the treatment of ectatic blood vessels found in port wine stains and in skin telangiectases. 23,24<br />

Treatment of cutaneous pigmentation has been achieved using melanin as the target chromophore. 25-27<br />

Selective targeting can also be applied to laser treatment of the pigmented trabecular meshwork cells.<br />

Latina and Park 28 demonstrated that a 532 nm Q-switched, frequency-doubled Nd:YAG laser with<br />

nanosecond pulse duration is capable of selective targeting of pigmented trabecular meshwork cells<br />

without collateral thermal damage to the adjacent non-pigmented trabecular meshwork cells and<br />

underlying trabecular beams. Based on this mechanism of selective targeting of pigmented trabecular<br />

meshwork cells, Selective Laser Trabeculoplasty, or SLT, is proposed as an alternative to ALT in the<br />

management of glaucoma.<br />

This report on performance data generated with the Coherent Selecta <strong>7000</strong> serves to establish substantial<br />

equivalence of this laser to predicate devices indicated for use in laser trabeculoplasty, and to<br />

demonstrate that the lower pulse energy and shorter pulse duration of the Coherent Selecta <strong>7000</strong><br />

results in selective destruction of pigmented trabecular meshwork cells, with minimal or no damage<br />

to non-pigmented cells and collagen beams of the trabecular meshwork, as compared to trabeculoplasty<br />

performed with the predicate devices.<br />

1.0 | Background and Rationale for SLT<br />

3


42.0<br />

| Coherent Selecta <strong>7000</strong><br />

Selective Laser Trabeculoplasty (SLT) is performed using the Coherent Selecta <strong>7000</strong>, a frequencydoubled,<br />

Q-switched Nd:YAG laser which delivers single laser pulses of 0.1 to 2.0 millijoules per<br />

pulse with a pulse duration of approximately 3 nanoseconds. This low energy, short pulsed laser<br />

treatment confines thermal damage to cells containing the target pigment or chromophore; in the<br />

case of trabecular meshwork, this target is represented by the pigmented cells. In addition to<br />

targeting the pigmented cells, the nanosecond pulse duration is a key factor in preventing collateral<br />

thermal and coagulative damage, since the pulse duration is less than the thermal relaxation time of<br />

1 microsecond for the melanin chromophore in the pigmented cells. The thermal relaxation time is<br />

the time required by the chromophore, in this case melanin, to convert the absorbed electromagnetic<br />

radiant energy into heat energy. When the deposition of radiant energy is rapid, as with a nanosecond<br />

pulse, only minimal conversion of radiant energy to heat occurs. Thermal dissipation to surrounding<br />

tissue is limited, as is coagulative damage, such that even in the treatment area, non-pigmented cells<br />

and structures are unaffected.


Preclinical evaluation of the Coherent Selecta <strong>7000</strong> laser has been conducted with the objective of<br />

demonstrating that this laser can selectively target pigmented trabecular meshwork cells while sparing<br />

adjacent, non-pigmented cells and tissues from coagulative damage. Additionally, testing was performed<br />

to establish the appropriate treatment parameters for selective targeting of pigmented trabecular<br />

meshwork cells and to evaluate the acute morphologic changes following selective laser trabeculoplasty.<br />

In vivo evaluation of SLT in cynomolgus monkeys was conducted to further establish the selective<br />

targeting of SLT in the ocular tissue, and to better define the underlying mechanism of action of SLT.<br />

3.1.1 In Vitro Studies to Determine Laser Effects in Trabecular Meshwork Cells<br />

(Thermal Effects of Laser Treatment)<br />

In a series of experiments conducted in pigmented and non-pigmented trabecular meshwork cells,<br />

three laser systems were used to evaluate threshold response, selected targeting of pigmented cells,<br />

and effects that are evident on transmission electron microscopy. 28 The laser systems were: (1) a<br />

continuous wave argon-ion laser; (2a) a flashlamp-pulsed dye laser with pulse duration of 8<br />

microseconds; (2b) a flashlamp-pulsed dye laser with pulse duration of 1 microsecond; (3a) a<br />

Q-switched frequency-doubled Nd:YAG laser emitting at 532 nm with a 10-nanosecond pulse duration,<br />

and (3b) a Q-switched normal mode Nd:YAG laser emitting at 1064 nm and with a 10-nanosecond<br />

pulse duration. Pigmented trabecular meshwork cells were generated by challenging confluent trabecular<br />

meshwork cell cultures with melanin.<br />

3.0 | Preclinical Studies<br />

Determination of threshold response was performed by irradiating cell cultures of pigmented and<br />

non-pigmented trabecular meshwork cells at various radiant exposures. Control cells were prepared<br />

by challenging non-pigmented trabecular meshwork cells with latex microspheres to determine the<br />

effect of laser irradiation on cells with particulate material but without the chromophore presented in<br />

the pigmented cells. The threshold radiant exposure was defined as the exposure where 50% cell<br />

cytoxicity was observed. Following determination of threshold radiant exposure, the three types of<br />

cell cultures were irradiated at radiant exposures ranging from sub-threshold to supra-threshold, to<br />

determine the range where selective targeting could be achieved. Cell cytoxicity and selectivity was<br />

determined using a fluorescent viability/cytotoxicity assay performed within 30 minutes of laser irradiation.<br />

Cells were also prepared for transmission electron microscopy (TEM) in order to determine the<br />

extent of cellular and intracellular damage, and to evaluate damage to adjacent non-pigmented cells.<br />

Following irradiation of the cell cultures with each of the laser systems evaluated, there was no evidence<br />

of cellular injury in the non-pigmented trabecular meshwork control cells and in the control cells<br />

with latex microspheres over a fluence of 10-10 6 mJ/cm 2 . Selective targeting of pigmented trabecular<br />

meshwork cells was achieved at threshold radiant exposures with the flashlamp pumped dye laser at<br />

588 nm with 1-microsecond pulse duration and the frequency-doubled Q-switched Nd:YAG laser at<br />

532 nm and 10-nanosecond pulse duration. With these two lasers, irradiation with fluences greater<br />

than 20 times the threshold exposure dose resulted in complete (i.e., 100%) cytotoxicity of pigmented<br />

5


| Preclinical Studies<br />

63.0<br />

trabecular meshwork cells. It was not possible to achieve selective targeting of only pigmented trabecular<br />

meshwork cells using either the continuous wave argon-ion laser emitting at 514 nm or the<br />

flashlamp-pulsed dye laser emitting at 590 nm with a 8-microsecond pulse. With these lasers, both<br />

pigmented and non-pigmented trabecular meshwork cells within the irradiation zone were non-selectively<br />

injured, and at fluences greater than threshold, many cells within the irradiation zone were<br />

vaporized. However, selective targeting of pigmented cells without injury to adjacent non-pigmented<br />

cells was achieved using pulse duration of 1 microsecond or less. The adjacent non-pigmented cells<br />

showed no evidence of cellular damage, and the damage to the pigmented cells was so subtle that<br />

these cells were difficult to differentiate morphologically from the other cells. This suggests that not<br />

only can pigmented cells be selectively targeted, but that this targeting can be accomplished without<br />

gross disruption of the cellular architecture.<br />

Transmission electron microscopy was used to assess the extent of cellular and/or intracellular damage,<br />

and the extent of damage to adjacent non-pigmented cells. Following irradiation with a 10-nanosecond<br />

pulse, intracellular damage was noted in pigmented cells, while there was no evidence of ultrastructural<br />

damage to adjacent non-pigmented cells, further supporting the confinement of laser damage only to<br />

pigmented cells.<br />

These findings established the ability of single laser pulses of short pulse duration and low fluence to<br />

selectively target pigmented trabecular meshwork cells in which melanin serves as the chromophore<br />

or target to absorb laser energy. In this study, large irradiation zones or spot diameters (ranging from<br />

200 µm to 2 mm) were utilized; this is in contrast to the smaller spot diameters (50 µm) utilized clinically<br />

in ALT procedures. Because tissue effects are localized at the subcellular level without collateral<br />

thermal coagulative damage, the irradiation zone can be of any shape. Additionally, the data generated<br />

in this study suggest that the appropriate laser parameters established in the trabecular meshwork cell<br />

culture system are equivalent to treatment parameters used clinically in ALT. Irradiation of pigmented,<br />

non-pigmented and control cells with an argon-ion laser emitting at 514 nm yielded threshold powers<br />

for cell damage consistent with treatment powers used clinically when performing ALT.<br />

3.1.2 In Vitro Studies to Evaluate Cytokine Responses to ALT and SLT (Biological Effects<br />

of Laser Treatment)<br />

The thermal effects of ALT elicit a healing process that includes cellular responses which can lead to<br />

either the improvement or deterioration of the function of the trabecular meshwork. For example,<br />

while macrophages are recruited to the area of themal damage for the trabecular meshwork clearance<br />

of pigment and debris that relieves obstruction in aqueous outflow, at the same time, trabecular cell<br />

division within the first few days after ALT 29 can counterbalance macrophage activity via scar formation,<br />

which can lead to eventual blockage of outflow. A series of studies have been undertaken to elucidate<br />

the cellular and biochemical responses to ALT that occur in the trabecular meshwork, and to compare<br />

these to the responses of this tissue to SLT, also at the cellular level.


Since it is known that metalloproteinases are involved in the turnover, remodeling and maintenance<br />

of trabecular extracellular matrix, 22 several groups of investigators examined the regulation of metalloproteases<br />

and their inhibitors. 30,31 To test the hypothesis that extracellular matrix turnover mediated<br />

by matrix metalloproteinases modulates aqueous humor outflow facility, perfused human anterior<br />

segment organ culture was utilized. 30 Purified matrix metalloproteinases, tissue inhibitors of metalloproteinases<br />

(TIMPs), and synthetic inhibitors of matrix metalloproteinases were added to the perfusion<br />

medium. In this model, addition of interleukin-1 alpha (Il-1 alpha) increased outflow facility, while<br />

inhibition of endogenous trabecular metalloproteinase activity using TIMP reduced outflow rates.<br />

These changes in outflow rates were reversible, with changes requiring 1 to 3 days, suggesting that<br />

modulation of matrix metalloproteinases and their inhibitors can be important in the regulation of<br />

aqueous humor outflow following laser trabeculoplasty.<br />

In another of these studies designed to examine modulation of trabecular meshwork matrix metalloproteinase<br />

expression, porcine trabecular meshwork cells were treated with several doses of various<br />

growth factors and cytokines, and the culture media was analyzed after 24, 48 and 72 hours. The<br />

most significant effects on trabecular matrix metalloproteinases were observed with tumor necrosis<br />

factor (TNF) alpha and beta, and interleukin-1 alpha. Other growth factors and cytokines elicited<br />

only small to negligible effects. 31 Thus, certain cytokines may play a role in trabecular cell regulation,<br />

thereby affecting aqueous humor outflow.<br />

3.0 | Preclinical Studies<br />

To further evaluate the factors that mediate matrix metalloproteinase expression, human anterior segment<br />

organ cultures were subjected to argon laser treatment (ALT) using standard clinical parameters, and<br />

then returned to culture. 32 The resulting culture medium was then applied to fresh anterior segment<br />

organ cultures, and was shown to induce typical trabecular cell division, as well as elevated levels of<br />

interleukin-1 beta (IL-1 beta) and tumor necrosis factor (TNF) alpha. Thus, ALT induced the expression<br />

and secretion of both of these cytokines, which in turn initiate remodeling of the juxtacanalicular<br />

extracellular matrix, restoring normal outflow facility.<br />

Alvarado and colleagues further expanded on these findings by evaluating the response of trabecular<br />

meshwork cells to both ALT and SLT, to assess the cellular response, and to evaluate cytokine induction<br />

in response to laser exposure. 33 Both the time course, as well as the magnitude of induction of the<br />

cytokine interleukin-1 alpha were evaluated following ALT and SLT, while induction of TNF-alpha<br />

and interleukin-1 beta were assessed after exposure to SLT. Both ALT and SLT treatment of cultured<br />

trabecular meshwork cells resulted in induction of interleukin-1 alpha, with a dose-dependent<br />

response observed following SLT performed with energy at 0.1 mJoule and at 1.0 mJoule. While minimal<br />

levels of intraleukin-1 alpha were induced with laser treatment at 0.1 mJoule, induction of this<br />

cytokine increased substantially with laser treatment of 1.0 mJoule, and with increasing concentrations<br />

of melanin in the cell culture. Following exposure to SLT, levels of TNF-alpha and interleukin-1<br />

beta were also increased in a dose-dependent fashion. Thus, with higher levels of laser energy, higher<br />

concentrations of these cytokines were generated by cultures of trabecular meshwork cells. These<br />

7


| Preclinical Studies<br />

83.0<br />

investigators postulate that the selective induction of interleukin-1 alpha by both ALT and SLT plays<br />

an important role in directing injured trabecular meshwork cells toward the established pattern of<br />

wound-healing activities. In addition to the effects of cytokines in initiating remodeling of the juxtacanalicular<br />

extracellular matrix, and thereby contributing to increased outflow facility, as described<br />

above, changes in the glycosaminoglycans of the trabecular meshwork, and the recruitment of<br />

macrophages following ALT and SLT may also be a result of interleukin-1 alpha induction. 33<br />

3.2 Ex Vivo Study (Human Globes)<br />

The acute morphologic changes induced by selective laser trabeculoplasty (SLT) were assessed using<br />

scanning and transmission electron microscopy, and were compared to the tissue effects induced by<br />

ALT using human eye bank eyes. 34 Half of the trabecular meshwork of each of 8 eye bank eyes was<br />

treated with ALT, and the other half with SLT, using the Coherent Selecta <strong>7000</strong> frequency-doubled<br />

Nd:YAG laser. Following laser treatment, specimens were submitted for evaluation by light<br />

microscopy, scanning electron microscopy (SEM) and transmission electron microscopy (TEM).<br />

In trabecular meshwork treated with ALT, scanning electron microscopy revealed crater formation in<br />

the uveal meshwork, with scrolling, whitening and bleb formation of the surrounding collagen, coagulative<br />

damage, and disruption of the endothelial cells. In contrast, SEM performed in SLT-treated<br />

trabecular meshwork revealed no evidence of crater formation, minimal evidence of mechanical<br />

damage, and intact uveal meshwork and corneal scleral trabecular sheets.<br />

Transmission electron microscopy (TEM) performed following ALT revealed disrupted trabecular<br />

meshwork, with fragmentation and fusion of the collagen framework and the trabecular endothelial<br />

cells. Similar evaluation of the trabecular meshwork following SLT revealed corneoscleral meshwork<br />

of normal appearance, with only minor disruptions in the pigmented meshwork cells.<br />

This morphologic comparison demonstrates that ALT causes disruption of the trabecular beams of<br />

the uveal and corneoscleral meshwork, with coagulative damage. In contrast, following SLT, there<br />

was no alteration of the collagen structure and central core regions of the trabecular beams in the<br />

uveal and corneoscleral meshwork.<br />

3.3.1 In Vivo Study (Primates) — Trabecular Meshwork Histology Following SLT<br />

The acute effects of SLT on the morphology of trabecular meshwork were evaluated in owl monkeys,<br />

a model chosen for the close resemblance of its trabecular meshwork structure and pigmentation to<br />

that of humans. 35 The SLT procedure was performed on the eyes of four primates, however half of<br />

each eye (either nasal or temporal 180°) was left untreated. The animals were sacrificed at 24 hours<br />

or 28 days, and tissue sections from each quadrant were processed for light and electron microscopy.<br />

Histologic evaluation of tissue of untreated tissue from the 24-hour sacrifice revealed compact, highly<br />

cellular trabecular meshwork with healthy cells lining the collagen beams, i.e., with normal morphology.<br />

Examination of tissue exposed to SLT revealed disrupted pigmented trabecular meshwork cells, and


cell debris, as well as pigment within the aqueous channels. However, the overall architecture of the<br />

trabecular meshwork, including the integrity of the inner wall of Schlemm’s canal, was well preserved<br />

without evidence of thermal damage. Similar histologic observations were made at 28 days. The<br />

trabecular meshwork structure was well preserved with an intact Schlemm's canal and healthyappearing<br />

non-pigmented trabecular meshwork cells lining the inner wall. Several engorged<br />

macrophages were present in the trabecular meshwork.<br />

The results of this histologic evaluation confirm the selective targeting of pigmented trabecular<br />

meshwork cells by SLT, and the absence of thermal damage to underlying tissue and structures in<br />

the trabecular meshwork.<br />

3.3.2 In Vivo Study (Primates) — Biological Effects of Laser Treatment<br />

Alvarado 13 has proposed that monocytes which normally circulate in small numbers in the aqueous<br />

humor are activated and transformed into macrophages upon interacting with injured tissues.<br />

Introduction of macrophages into the anterior chamber results in a rapid and persistent increase in<br />

outflow facility, 13 while the presence of dead macrophages has been associated with a marked obstruction<br />

of aqueous outflow. 36 Following ALT, numerous macrophages are observed in the trabecular<br />

meshwork; these cells engulf melanin granules, reach an enormous size and clear the pigment granules<br />

from the trabecular meshwork via Schlemm’s canal. In cynomolgous monkeys treated with SLT, the<br />

number of monocytes and macrophages was found to be 5- to 8-fold higher than in the untreated<br />

controls. These findings suggest that injury to pigmented trabecular meshwork cells induced by SLT<br />

results in the release of factors and chemoattractants which recruit macrophages in a manner similar<br />

to that previously observed for ALT. This recruitment of macrophages by both ALT and SLT may be at<br />

least in part responsible for the increased aqueous outflow facility observed following these procedures.<br />

3.0 | Preclinical Studies<br />

Scanning electron microscopy of the trabecular meshwork tissues in the cynomolgous monkeys treated<br />

with SLT revealed no localized effects of treatment, such that treated and untreated tissues were indistinguishable<br />

from each other.<br />

3.4 Summary of Preclinical Studies<br />

The extensive preclinical investigations summarized herein serve to establish the ability of the Q-switched,<br />

frequency-doubled Nd:YAG laser, emitting at 532 nm with pulse duration in the nanosecond range,<br />

to selectively target pigmented trabecular meshwork cells, utilizing melanin as the target chromophore.<br />

By selectively targeting pigmented cells, thermal damage to surrounding cells and tissue was avoided.<br />

These observations are consistent with previously reported data on selective photothermolysis, in<br />

which the use of a short laser exposure confines thermal damage to the target (i.e., melanin), if it is<br />

equal to or shorter than the thermal relaxation time of the target. 37 With the thermal relaxation time<br />

for melanin at approximately 1 microsecond, pulses with duration of less than 1 microsecond deposit<br />

energy within the target more rapidly than this energy can diffuse away. While the temperature within<br />

9


3.0 | Preclinical Studies<br />

the target, in this case pigmented trabecular meshwork cells, increases to levels adequate to cause<br />

selective cellular destruction, thermal diffusion to surrounding non-pigmented cells is minimized.<br />

This selective targeting of pigmented cells was reflected in the morphologic evaluation of both irradiated<br />

cell cultures of trabecular meshwork cells, and actual trabecular meshwork from human eye bank<br />

eyes. In both cases, changes in cellular ultrastructure were observed only in pigmented trabecular<br />

meshwork cells, while adjacent non-pigmented cells and tissues were spared. This was confirmed in<br />

primate eyes, in which SEM revealed no localized effects following SLT. This was in contrast to the<br />

tissue exposed to ALT, in which non-specific destruction of trabecular meshwork cells, and thermal<br />

damage to underlying and adjacent collagen beams were observed. Additionally, the significant<br />

increase of macrophages in primate eyes following SLT, as has been reported following ALT, confirms<br />

the contribution of biologic effects to the usefulness of both of these procedures.<br />

10


In addition to the definitive body of preclinical data establishing the effect of SLT on the trabecular<br />

meshwork, extensive supportive clinical data have been generated on the SLT procedure. Clinical<br />

experience with SLT gained from evaluation of over 1,100 eyes treated in prospective clinical studies<br />

around the world supports both the safety and effectiveness of this procedure as an alternative to ALT.<br />

This clinical experience with the Coherent Selecta <strong>7000</strong> laser for laser trabeculoplasty is summarized<br />

in a table on the following pages. As shown consistently in the summary information, reduction of<br />

intraocular pressure following SLT was very similar to the decrease in IOP reported for eyes that<br />

underwent ALT in the control arms of these studies. And while the number of eyes in the ALT control<br />

groups was relatively small, particularly in comparison to the large number of eyes treated with SLT,<br />

the observations reported in this population of eyes were consistent with the extensive published<br />

literature on ALT, with regard to both intraocular pressure reduction and safety of the procedure.<br />

More detailed information on several of the key trials follows the table of worldwide clinical studies<br />

of the Coherent Selecta <strong>7000</strong> laser used to perform selective laser trabeculoplasty, or SLT.<br />

4.0 | Supportive Information — Clinical Experience with SLT<br />

11


124.0 | Supportive Information — Clinical Experience with SL T<br />

Table 1A Worldwide Clinical Studies of the Selective Laser Trabeculoplasty (SLT)<br />

Author Publication Study Design Number Duration Baseline IOP IOP at Decrease Other Comment<br />

of Patients of Follow-up (mean, S.D.) Follow-up in IOP Outcomes<br />

(mean, S.D.)<br />

Anschutz - Prospective, 398 12–24 months 22.3 mmHg 17.9 4.4 mmHg 38% of eyes 82% of<br />

single arm (12 months) (19.8%) required no patients<br />

18.2 (12 months) glaucoma responded<br />

(24 months) 4.1 mmHg medication, with IOP<br />

(18.4%) 52% reduced reduction<br />

(24 months) the use of ≥3 mmHg<br />

glaucoma<br />

medication<br />

Damji et al. British Jour Prospective, ALT - 42 6 months 23.6 mmHg ALT: ALT: – No change in<br />

Ophthalmol randomized SLT - 35 for both groups 18.3 mmHg 5.0 mmHg IOP in<br />

1999;83: SLT vs ALT SLT: SLT: untreated<br />

718–722 38 18.0 mmHg 5.2 mmHg fellow eyes<br />

(20%)<br />

Garza-Saide – Prospective, 24 Average 27.6 mmHg 18.2 mmHg 9.4 mmHg Transient –<br />

et al. feasibility, 32 weeks (33%) increase in IOP<br />

2 clinical sites postprocedure<br />

in 7 eyes (24%)<br />

Goulas et al. – Non-randomized ALT - 159 3 years ALT: ALT: ALT: 27% SLT group<br />

comparison of SLT - 42 24.3 mmHg 17.8 mmHg SLT: 28% had significant<br />

ALT and SLT (S.D. 4.2) (S.D. 3.1) decrease in<br />

(subgroup of SLT: SLT: use of<br />

study under 23.9 mmHg 17.2 mmHg glaucoma drugs;<br />

G960194) (S.D. 4.2) (S.D. 3.9) no effect in<br />

ALT group –<br />

Hong – Prospective, 20 3 months 20.5 mmHg 14.2 mmHg 30%<br />

single arm study – –


4.0 | Supportive Information — Clinical Experience with SLT<br />

13<br />

Worldwide Clinical Studies of the Selective Laser Trabeculoplasty (SLT) cont’d<br />

Author Publication Study Design Number Duration Baseline IOP IOP at Decrease Other Comment<br />

of Patients of Follow-up (mean, S.D.) Follow-up in IOP Outcomes<br />

(mean, S.D.)<br />

Kano et al. Nippon Ganka Prospective, 67 6 months 22.4 mmHg 18.0 mmHg 20% Transient increase –<br />

Gakkai Zasshi single arm study in IOP in 25%<br />

1999;103: of cases<br />

612–616 39<br />

Table 1A<br />

Kaulen et al. – Prospective, 221 12 months 24.7 mmHg 17.9 mmHg 23% – –<br />

single arm study<br />

Kim et al. Ophthalmic Prospective, 13 pts 1 year 24.4 mmHg 19.5 mmHg 20% Transient increase –<br />

Surgery & Lasers single arm study (16 eyes) in IOP in 13%<br />

2000;31: of cases, managed<br />

394–399 40 with apraclonidine<br />

Lachkar et al. – Prospective, 24 eyes 5 weeks 24.6 mmHg 17.7 mmHg 28% Transient increase –<br />

single arm study<br />

in IOP in 2 eyes<br />

Lanzetta et al. British Journal Prospective, 8 6 weeks 26.6 mmHg 16 mmHg 40% One eye with No significant<br />

of Ophthalmol single arm study (S.D. 7) (S.D. 2.6) a transient anterior<br />

1999;83:29–32 41 increase in IOP chamber<br />

cell/flare<br />

Latina et al. Ophthalmol Prospective, 125 26 weeks 25.3 mmHg 20.9 mmHg 17% 9 eyes (7%) Cell/flare<br />

1998;105: single arm study had transient transient,<br />

2082–2090 42 increased IOP minimal/mild<br />

Rozsival et al. – Prospective, 171 Average – – Decrease IOP No significant Transient,<br />

single arm study 6 months >3 mmHg in postprocedure mild AC cells<br />

146 eyes (85%) increase in IOP and flare<br />

13


144.0 | Supportive Information — Clinical Experience with SL T<br />

4.1 A Prospective, Multicenter Clinical Trial — Frequency Doubled Nd:YAG Ophthalmic<br />

Laser for Selective Laser Trabeculoplasty<br />

4.1.1 Methods<br />

A multicenter, prospective clinical study was conducted by Coherent Medical Group under IDE<br />

G960194 to evaluate the Coherent Selecta <strong>7000</strong> for use in performing laser trabeculoplasty (SLT) to<br />

reduce intraocular pressure in patients with open-angle glaucoma. A total of 125 patients were<br />

enrolled at four clinical sites. The study population included patients with uncontrolled open-angle<br />

glaucoma who were on maximum tolerated medical drug therapy (Max Rx group) as well as patients<br />

who had previously failed laser trabeculoplasty (PFLT group). Treatment with the Coherent Selecta <strong>7000</strong><br />

was performed over 180° of the trabecular meshwork, and patients were then followed for 26 weeks.<br />

4.1.2 Results<br />

4.1.2.1 Demographics and Baseline Information<br />

Of the 125 patients enrolled in the study, data were available for 122 patients and 120 of the 122<br />

patients underwent SLT; two patients did not meet the entry criteria and therefore were not treated.<br />

Demographic information, as well as information on patient status at entry into the study are shown<br />

in Table 1.<br />

Table 1: Demographic Information and Baseline Characteristics (N=120)<br />

Age (years) 67.7(S.D. 10.7; range 29 to 90)<br />

Sex (% female) 65 (54.2%)<br />

Race<br />

Caucasian 77 (64.2%)<br />

Black 34 (28.3%)<br />

Hispanic 9 (7.5%)<br />

Diagnosis*<br />

Primary open-angle 99 (82.5%)<br />

Pseudoexfoliative 7 (5.8%)<br />

Pigmentary 7 (5.8%)<br />

Other 9 (7.3%)<br />

*Several patients had multiple diagnoses.<br />

The majority of patients enrolled (82.5%) presented with primary open-angle glaucoma. Other<br />

diagnoses included pseudoexfoliative glaucoma, pigmentary glaucoma, mixed mechanism glaucoma,<br />

open-angle glaucoma in pseudophakia, and juvenile open-angle glaucoma; several patients had<br />

multiple diagnoses, including a patient with both pigmentary and pseudoexfoliative glaucoma.


4.1.2.2 Intraocular Pressure Reduction<br />

At 26 weeks post-SLT, a significant decrease in intraocular pressure as compared to baseline IOP was<br />

observed, with a mean decrease in IOP of 4.4 mmHg for the overall study population. Patients with<br />

higher intraocular pressure at baseline experienced a larger decrease in IOP than those patients with<br />

lower initial IOP. A small decrease in IOP was also observed in untreated fellow eyes, however the<br />

magnitude of change from baseline in fellow eyes was insignificant when compared to the significant<br />

improvement in IOP in the SLT-treated eyes (Tables 2 and 3).<br />

Table 2: Mean Intraocular Pressure (mmHg) SLT-Treated Eyes and Fellow Eyes (N=120)<br />

SLT-Treated Eyes<br />

Fellow Eyes (Untreated)<br />

(mean, S.D.)<br />

(mean, S.D.)<br />

Baseline 25.3 mmHg (2.9) 21.4 mmHg (4.1)<br />

Day 1 18.2 mmHg (4.4) 20.6 mmHg (4.7)<br />

Week 26 20.9 mmHg (3.6) 19.5 mmHg (4.9)<br />

Examination of mean IOP reduction, expressed in mmHg and as a percent of the baseline intraocular<br />

pressure, serves to further emphasize the significant improvement in intraocular pressure in the SLTtreated<br />

eyes. In contrast, fellow eyes experienced only a small decrease in IOP over the course of the<br />

study.<br />

Table 3: Intraocular Pressure Reduction (mmHg) SLT-Treated Eyes and Fellow Eyes (N=120)<br />

SLT-Treated Eyes<br />

Fellow Eyes (Untreated)<br />

(mean, S.D.)<br />

(mean, S.D.)<br />

Day 1 7.1 mmHg (27.9%) 0.9 mmHg (3.5%)<br />

Week 26 4.4 mmHg (17.1%) 1.9 mmHg (8.3%)<br />

The decrease in intraocular pressure in the SLT-treated eyes was nearly 10-fold the decrease observed<br />

in the untreated fellow eyes at Day 1, and twice as great as the decrease in fellow eyes at Week 26.<br />

4.1.2.3 Safety<br />

Primary measures of safety in this study included visual acuity, cup to disc ratio, results of anterior<br />

segment examination, and adverse events. No changes were observed in either visual acuity or cup to<br />

disc ratio from baseline to 26 weeks. Anterior segment findings included minimal cells and flare in<br />

the SLT-treated eyes on postoperative Day 1. A mean score of 1, on a scale of 0 to 4, was reported for<br />

the first two hours following SLT. This had decreased to a mean of 0.3 on the first postoperative day,<br />

and had resolved by one week. Mean flare was 0.2, and no further cells or flare were noted for the<br />

remainder of the study. Thus, cells and flare were minimal, and limited to the immediate postoperative<br />

period. It should be noted that even though the presence of cells and flare in the anterior chamber is<br />

expected following laser treatment of the trabecular meshwork, all observations of cells and flare were<br />

reported by the study investigators as adverse events.<br />

4.0 | Supportive Information — Clinical Experience with SLT<br />

15


164.0 | Supportive Information — Clinical Experience with SL T<br />

An increase in IOP of ≥ 10 mmHg above the baseline intraocular pressure was reported in 9 (7.5%)<br />

of the SLT-treated eyes. This was considered likely to have been related to the study treatment (SLT)<br />

in 7 of the 9 eyes. In 5 of these patients, the increase in IOP occurred within the first 24 hours following<br />

SLT, and resolved within 1 day. In the other two patients, the increase in IOP occurred within<br />

1 week of treatment and resolved within 2 weeks. In one case, a later occurrence of increased IOP<br />

required surgical intervention (trabeculectomy), however this was considered by the study investigator<br />

to be unrelated to SLT treatment. It should be noted that study patients were not pretreated with<br />

apraclonidine 1%, even though this is the current standard of care for patients undergoing ALT,<br />

with the goal of avoiding a laser-induced pressure spike.<br />

Other reported complications that were considered to be related to treatment included conjunctivitis,<br />

eye pain, blurred vision, iritis, corneal edema, corneal lesion, and headache.<br />

4.1.3 Summary and Conclusions<br />

In this prospective, multicenter clinical trial, 120 patients with uncontrolled open-angle glaucoma<br />

were successfully treated with Selective Laser Trabeculoplasty, as an alternative to argon laser trabeculoplasty<br />

(ALT). Significant reduction in intraocular pressure was observed in the SLT-treated eyes over<br />

the course of the 26-week follow-up, and the safety profile was consistent with that reported for ALT. 4,5<br />

The protocol for this clinical trial of SLT described use of a historical control ALT population, derived<br />

from the published literature on results of ALT. However, the historical control developed for use as a<br />

comparator against SLT proved to be invalid for several critical reasons. First, the historical control<br />

population proved not to be matched to the SLT study population with regard to prior glaucoma<br />

treatment. In contrast to the study population, which consisted of patients with uncontrolled openangle<br />

glaucoma who were on maximum tolerated medical drug therapy (Max Rx group) as well as<br />

patients who had previously failed laser trabeculoplasty (PFLT group), the historical control population<br />

included newly-diagnosed primary open-angle glaucoma. There was no attempt to match the<br />

historical control population to the study population with respect to concomitant glaucoma medications.<br />

Of even greater consequence, the historical control population excluded patients who had previously<br />

failed laser treatment, a group that constituted over 50% of the patients in the SLT study, a<br />

group that typically responds poorly to laser trabeculoplasty retreatment. And in some instances the<br />

control group excluded results of ALT failures, further biasing the derived IOP mean. In addition to<br />

inadequate matching of baseline characteristics, the historical control population included patients<br />

who underwent ALT performed on 360° of the trabecular meshwork, while SLT was limited to only<br />

180° such that even the extent of treatment was not comparable for the two populations. Thus,<br />

based on inadequate matching of the historical control population to the SLT population, use of this<br />

population as a control was invalidated.


In summary, in this clinical trial of Selective Laser Trabeculoplasty, the intraocular pressure-reducing<br />

effect of SLT was demonstrated in a population of glaucoma patients, with a safety profile similar to<br />

that of ALT.<br />

4.2 A Prospective, Randomized, Clinical Trial Comparing the Efficacy of Selective Laser<br />

Trabeculoplasty (SLT) to Argon Laser Trabeculoplasty (ALT)<br />

4.2.1 Methods<br />

In this prospective clinical trial, patients with uncontrolled open-angle glaucoma (OAG), including<br />

primary, pigmentary and pseudoexfoliative forms of OAG, were recruited from two glaucoma practices<br />

located in Ottawa, Canada (K. Damji, M.D., University of Ottawa; W. Rock, M.D., private practice,<br />

Ottawa). After being screened for eligibility, 77 patients were randomly assigned to undergo either<br />

Selective Laser Trabeculoplasty (SLT) or Argon Laser Trabeculoplasty (ALT), performed at the<br />

University of Ottawa by one of these two investigators. The inferior 180° of the angle was generally<br />

treated, unless the patient had previously undergone ALT, in which case the superior 180° was treated<br />

with the randomly assigned laser treatment, i.e., SLT or ALT. Following the laser procedure, patients<br />

were examined at one hour, to assess intraocular pressure and anterior chamber reaction, and then at<br />

1 week, and 1, 3, and 6 months.<br />

4.2.2 Results<br />

4.2.2.1 Demographics and Baseline Characteristics<br />

Demographic information as well as information on diagnosis and baseline intraocular pressure are<br />

shown in Table 4 for the total study population of 77 patients.<br />

The study groups were very similar with regard to age and sex, and the majority (approximately<br />

60%) had an initial diagnosis of primary open-angle glaucoma. The SLT treatment group had a history<br />

of more drug use, with a mean of 3.6 glaucoma medications utilized as compared to 2.9 medications<br />

in the ALT group (p=0.04). Baseline intraocular pressure was identical for the two groups, at 23.6 mmHg.<br />

4.0 | Supportive Information — Clinical Experience with SLT<br />

17


184.0 | Supportive Information — Clinical Experience with SL T<br />

Table: 4 Demographic and Baseline Characteristics by Treatment Group<br />

ALT Group<br />

SLT Group<br />

Characteristic (n=42) (n=35)<br />

Age (years) 70.1 (10.2) 66.2 (10.4)<br />

Sex (% female) 50.0 (21/42) 51.4 (18/35)<br />

Diagnosis<br />

Primary open-angle 59.5 (25/42) 57.1 (20/35)<br />

Pseudoexfoliative 28.6 (12/42) 20.0 (7/35)<br />

Pigmentary 2.4 (1/42) 8.6 (3/35)<br />

Other 9.5 (4/42) 14.3 (5/35)<br />

No. of Glaucoma Medications 2.9 (1.4) 3.6 (1.9)<br />

IOP (mm Hg) 23.6 (3.5) 23.6 (4.1)<br />

4.2.2.2 Intraocular Pressure Reduction<br />

As shown in Table 5, a comparison of mean IOP values between ALT and SLT-treated groups at each<br />

time point, unadjusted for the preoperative IOP level, revealed no differences between groups at any<br />

of the time points from preoperative to Month 6. Baseline mean intraocular pressure was identical for<br />

the two groups (23.6 mmHg), and remained similar over the 6-month follow-up period (18 mmHg<br />

at 6 months).<br />

Table 5: Mean Intraocular Pressure (mmHg) by Treatment Group and Time<br />

ALT Group<br />

SLT Group<br />

Time N Mean (S.D.) N Mean (S.D.)<br />

Baseline 42 23.6 (3.5) 35 23.6 (4.1)<br />

Day 7 39 20.1 (5.1) 32 21.3 (5.0)<br />

Month 1 38 19.7 (4.3) 33 20.6 (4.7)<br />

Month 3 35 20.2 (5.4) 32 20.1 (5.2)<br />

Month 6 34 18.3 (4.8) 30 18.0 (4.6)<br />

Mean reduction in IOP from the preoperative examination to post-treatment timepoints for the overall<br />

study group is shown in Table 6. As for mean intraocular pressure, no significant differences were<br />

observed between ALT and SLT-treated groups at any of the postoperative times. By Month 6, the<br />

average IOP reduction in both groups was at or above 5.0 mmHg.


Table 6: Mean Intraocular Pressure Reduction (mmHg) from the Preoperative Examination by<br />

Treatment Group and Time<br />

ALT Group<br />

SLT Group<br />

Time N Mean (S.D.) N Mean (S.D.)<br />

Day 7 39 3.7 (4.8) 32 2.2 (5.2)<br />

Month 1 38 4.0 (4.4) 33 3.1 (5.0)<br />

Month 3 35 3.2 (5.1) 32 3.1 (5.1)<br />

Month 6 34 5.0 (5.2) 30 5.2 (3.9)<br />

Percentage reduction from baseline is shown for the overall study group in Table 7. Consistent with<br />

the mean IOP reduction shown in Table 6, the mean percentage reduction in IOP from the preoperative<br />

examination to post-treatment time points did not differ substantially between groups. By Month<br />

6, percentage reductions in both ALT-treated and SLT-treated patients exceeded 20% from the preoperative<br />

baseline value.<br />

Table 7: Mean Intraocular Pressure Percentage Reduction from the Preoperative Examination by<br />

Treatment Group and Time<br />

ALT Group<br />

SLT Group<br />

Time N Mean (S.D.) N Mean (S.D.)<br />

Day 7 39 15.1 (17.9) 32 8.1 (21.2)<br />

Month 1 38 16.2 (16.9) 33 12.0 (18.6)<br />

Month 3 35 13.2 (20.3) 32 13.0 (20.9)<br />

Month 6 34 20.4 (20.5) 30 22.3 (16.0)<br />

Intraocular pressure in untreated fellow eyes was followed in a subgroup of patients who had previously<br />

failed laser treatment (PFLT subgroup), and is summarized in Table 8. In fellow eyes of patients<br />

in the ALT treatment group, a decrease was observed in mean IOP from baseline to Month 1, after<br />

which IOP remained largely unchanged through Month 6. In the SLT treatment group, IOP in fellow<br />

eyes remained fairly constant through the course of the 6-month follow-up.<br />

4.0 | Supportive Information – Clinical Experience with SLT<br />

19


204.0 | Supportive Information — Clinical Experience with SL T<br />

Table 8: Mean Intraocular Pressure in Untreated Fellow Eyes of the PFLT Subgroup by Treatment Group<br />

and Time<br />

ALT Group<br />

SLT Group<br />

Time N Mean (SD) N Mean (SD)<br />

Baseline 17 21.4 (4.7) 10 17.9 (3.8)<br />

Month 1 16 17.5 (5.2) 10 17.4 (3.4)<br />

Month 3 12 18.5 (4.5) 9 16.2 (3.2)<br />

Month 6 16 16.8 (4.1) 9 16.4 (2.9)<br />

4.2.2.3 Safety<br />

Inflammatory changes (grading of anterior chamber cells and flare) at one hour and one week after<br />

laser treatment are shown in Table 9 or the total study population. One hour after SLT, there were<br />

significantly more cells in the anterior chamber than were reported for the ALT-treated group. A grading<br />

of 2+ or more was observed in 42.3% (n=11) of the SLT-treated group, versus 14.7% (n=5) of the<br />

ALT-treated group (P=0.02, Chi-square test). However, by one week after laser treatment, and continuing<br />

through the duration of follow-up, both treatment groups were essentially free of any signs of<br />

cells and flare in the anterior chamber.<br />

Table 9: Inflammatory Changes at One Hour and One Week Post-Treatment by Treatment Group and Time<br />

One Hour after Laser Treatment:<br />

Grading of Cells at One Hour<br />

N Zero One Two Three<br />

ALT 34 35.3 (12) 50.0 (17) 14.7 (5) 0.0 (0)<br />

SLT 26 11.5 (3) 46.2 (12) 26.9 (7) 15.4 (4)<br />

Grading of Flare at One Hour<br />

N Zero One Two Three<br />

ALT 33 45.5 (15) 45.5 (15) 9.1 (3) 0.0 (0)<br />

SLT 26 26.9 (7) 57.7 (15) 15.4 (4) 0.0 (0)


One Week after Laser Treatment:<br />

Grading of Cells at One Week<br />

N Zero One Two Three<br />

ALT 32 100.0 (32) 0.0 (0) 0.0 (0) 0.0 (0)<br />

SLT 26 100.0 (26) 0.0 (0) 0.0 (0) 0.0 (0)<br />

Grading of Flare at One Week<br />

N Zero One Two Three<br />

ALT 32 100.0 (32) 0.0 (0) 0.0 (0) 0.0 (0)<br />

SLT 25 96.0 (24) 4.0 (1) 0.0 (0) 0.0 (0)<br />

No serious or unanticipated adverse device effects were reported during this study. As a result of<br />

standard pretreatment with apraclonidine 1%, to prevent increases in intraocular pressure, two<br />

patients in each treatment group experienced an IOP spike of 5 mmHg or greater in the immediate<br />

post-treatment period, and there were no reports of peripheral anterior synechia.<br />

4.2.3 Summary and Conclusions<br />

In this randomized clinical trial of the effect of ALT and SLT on IOP reduction in patients with<br />

uncontrolled open-angle glaucoma, no overall treatment differences were observed at any postoperative<br />

time point through 6 months after the laser procedure. IOP reduction from the preoperative<br />

examination was comparable in both groups, and exceeded 5 mmHg by 6 months after surgery. The<br />

percentage reduction in IOP was also comparable in both groups, with a greater than 20% reduction<br />

from baseline IOP at 6 months after surgery.<br />

An increased inflammatory response was observed following SLT, with a significantly higher grade for<br />

anterior chamber cells at one-hour post-laser procedure than was reported for ALT. However, from<br />

one week post-treatment through the 6-month examination, the two groups were essentially free of<br />

any signs of cells or flare in the anterior chamber. No patients in the study developed peripheral<br />

anterior synechiae, and there were no complications or adverse events.<br />

These findings demonstrate that treatment with SLT results in a clinically substantial reduction in<br />

IOP 6 months after treatment, with a magnitude of IOP reduction in SLT treated eyes similar to that<br />

observed in ALT treated eyes.<br />

4.3 Summary of Clinical Experience with SLT<br />

4.0 | Supportive Information — Clinical Experience with SLT<br />

Clinical evaluation of SLT in over 1,100 eyes worldwide serves to establish the safety and effectiveness<br />

of this procedure as an alternative to ALT. Reduction of intraocular pressure following SLT in each of<br />

the studies reported was very similar to the decrease in IOP reported for ALT. This was consistent<br />

across studies in which there was a concurrent ALT control, as well as with the extensive body of<br />

21


4.0 | Supportive Information — Clinical Experience with SLT<br />

published data on ALT, demonstrating the comparable effects of both of these laser trabeculoplasty<br />

procedures.<br />

Safety outcomes following SLT were also consistent with the published safety data on ALT, with<br />

regard to all major safety parameters, including acute intraocular pressure increase and anterior<br />

chamber inflammation. Transient elevation of IOP was reported in 7% to 25% of eyes following SLT,<br />

varying by study and patient population. This compares favorably with published reports showing an<br />

increase in IOP of 5 mmHg or more in up to 33% of eyes, and an increase of some magnitude in up<br />

to 70% of eyes following ALT. 43-45 This prevalence of IOP rise following argon laser trabeculoplasty led<br />

to evaluation, and adoption of apraclonidine, administered one hour prior to treatment, as a standard<br />

of care in this patient population. 46 Published reports on anterior chamber reaction (cell and flare) are<br />

also consistent with the clinical data reported for SLT. 43<br />

In summary, the extensive body of clinical data on selective laser trabeculoplasty serves to support<br />

both the safety and effectiveness of this procedure, as well as the substantial equivalence of SLT to ALT.<br />

22


Ophthalmic lasers for use in laser trabeculoplasty are Class II devices, classified as per CFR 886.4390<br />

(product code HQF – laser, ophthalmic) and CFR 878.4810 (product code GEX – laser surgical<br />

instrument). Additionally, under CFR 886.4392, Nd:YAG lasers are identified as mode-locked or<br />

Q-switched solid state lasers which generate short pulse, low energy, high power radiation.<br />

The Coherent Selecta <strong>7000</strong> is substantially equivalent to commercially available lasers with regard to<br />

indication for use and system characteristics, as follows:<br />

- K913127 – Coherent Novus 3000 Argon Photocoagulator<br />

- K972514 – Laserex LP1532 Nd:YAG Photocoagulator<br />

- K962592 – Alcon Ophthalas 532 Solid State Photocoagulator<br />

- K900199 – Lasag Microruptor 2 Nd:YAG Ophthalmic Laser<br />

Like the Coherent Selecta <strong>7000</strong>, these predicate devices are indicated for use in laser trabeculoplasty,<br />

performed in patients with open-angle glaucoma. Additionally, the system characteristics of the<br />

Coherent Selecta <strong>7000</strong> and the predicate lasers are very similar, as shown in the table of technological<br />

comparison on the following page. With the exception of the Coherent Novus 3000 Argon<br />

Photocoagulator, the three other laser predicate devices utilize a Nd:YAG laser source and the Laserex<br />

and Alcon lasers emit at the same wavelength, i.e., 532 nm, as the Coherent Selecta <strong>7000</strong>. The<br />

Coherent Selecta <strong>7000</strong> and the predicate lasers employ a diode or HeNe aiming beam.<br />

The shorter pulse duration (i.e., 3 nanoseconds) of the Coherent Selecta <strong>7000</strong> is integral to the mechanism<br />

of selective targeting of pigmented trabecular meshwork cells, since this pulse duration is less<br />

than the thermal relaxation time of 1 microsecond for melanin. Pulses with duration of less than 1<br />

microsecond deposit energy within the target cells more rapidly than this energy can diffuse away.<br />

While the temperature within the target, i.e., pigmented trabecular meshwork cells, increases to levels<br />

adequate to cause selective cellular destruction, thermal diffusion to surrounding non-pigmented cells<br />

is minimized. Preclinical investigations in cultures of trabecular meshwork cells and in trabecular<br />

meshwork obtained from human eye bank eyes substantiated this mechanism of selective targeting of<br />

cells. Following SLT, changes in cellular ultrastructure were limited to pigmented trabecular meshwork<br />

cells, while adjacent non-pigmented cells and tissues were spared. This was in contrast to the<br />

tissue exposed to ALT, in which non-specific destruction of trabecular meshwork cells, and thermal<br />

damage to underlying and adjacent collagen beams were observed. These changes may cause or contribute<br />

to the elevated post-treatment intraocular pressure observed following ALT, and to the limited<br />

success of ALT retreatment. On this basis, it can be concluded that the shorter pulse duration and<br />

low energy of the Coherent Selecta <strong>7000</strong> are associated with less tissue damage than the predicate<br />

devices when used to perform laser trabeculoplasty. The Coherent Selecta <strong>7000</strong> thus offers an<br />

improved safety profile as compared to the predicate devices by providing greater protection to the<br />

trabecular meshwork and preserving this tissue for the possibility of additional medical, laser or surgical<br />

treatment. This is supported by clinical data demonstrating a similar effect of SLT and ALT on<br />

reduction of intraocular pressure and on patient safety.<br />

5.0 | Premarket Notification for the Selecta <strong>7000</strong> Laser<br />

23


245.0<br />

| Premarket Notification for the Selecta <strong>7000</strong> Laser<br />

Comparison of Coherent Selecta <strong>7000</strong> and Predicate Devices<br />

Coherent Selecta <strong>7000</strong> Coherent Novus 3000 Laserex LP1532 Alcon Ophthalas 532 Lasag Microruptor 2<br />

Nd:YAG Ophthalmic Laser Argon Ophthalmic Laser Nd:YAG Photocoagulator Solid State Photocoagulator Nd:YAG Ophthalmic Laser<br />

K913127 K972514 K962592 K900199<br />

Indications for Use Indicated for use in laser Indicated for use in the Indicated for use in laser Indicated for use in laser g<br />

Indicated for performing<br />

trabeculoplasty treatment of ocular trabeculoplasty trabeculoplasty the trabeculoplasty<br />

pathology including<br />

trabeculoplasty in<br />

Power/Energy 0.1–2.0 mJ 50–1750 mW 30 mW–2000 mW 50 mW–3000 mW 0.3–15.0 J<br />

Pulse Duration 3 ns 0.01–1.0 sec 0.01–2.0 sec 0.01–2.0 sec, and continuous 20 msec<br />

Laser Media Nd:YAG Argon Nd:YAG Nd:YAG Nd:YAG<br />

Wavelength 532 nm 488–514 nm (blue-green) 532 nm 532 nm 1064 nm<br />

Mode of Operation Q-Switched CW Q-switched CW<br />

procedure in patients with<br />

uncontrolled open-angle<br />

open-angle glaucoma<br />

Spot Size 400 µm 50–1000 µm 50–500 µm 50–1000 µm 70 µm<br />

Aiming Beam Diode, variable intensity Diode, variable intensity Diode, variable intensity HeNe, four intensities HeNe, maximum intensity<br />

from barely visible to from barely visible to from 0.05–0.95 mW selectable up to 1mW of 0.1 mW<br />

< 1.0 mW 1.5mW<br />

Laser Control System Microprocessor Microprocessor Microprocessor Microprocessor Microprocessor<br />

Cooling System Air cooled Air cooled Air cooled Internal water cooled Air cooled<br />

Slit Lamp Coherent LDS-10C Slit Lamp Coherent, Zeiss, Haag Streit Zeiss, Haag Streit Zeiss, Haag Streit Zeiss<br />

Dimensions Table: 94 cm x 58 cm 47 cm x 20 cm x 60 cm 19 cm x 31 cm x 51 cm 80 cm x 36 cm x 81 cm<br />

(37″ x 23″) (18.5″ x 8″ x 24″) (7.5″ x 12″ x 20″) (31.5″ x 14″ x 32″)<br />

Height: 152 cm (h x w x d) (h x w x d) (h x w x d,<br />

(60″) laser console only)<br />

Weight 123 kg (270 lbs) 30 kg (65 lbs) 17 kg (37 lbs) 99 kg (218 lbs) 350 kg (772 lbs)<br />

Electrical 115/230 VAC; 50/60 Hz; 100–120 VAC; 15–20 A 100–120 VAC, 220 VAC, 16 A, 50–60 Hz 240/220/210 V<br />

Requirements 4/2 A 220–240 VAC; 10 A 220–250 VAC 120/110/100 V<br />

50/60 Hz single phase, 50/60 Hz 50/60 Hz, 10/20 A


Bibliography<br />

1. Worthen DM, Wickham MG. Laser trabeculotomy in monkeys. Invest Ophthalmol Vis Sci 1973;12:707-711.<br />

2. Worthen DM, Wickham MG. Argon laser trabeculotomy. Trans Am Acad Ophthalmol Otolaryngol<br />

1974;78:371-375.<br />

3. Wise JB, Witter SL. Argon laser therapy for open-angle glaucoma: A pilot study. Arch Ophthalmol<br />

1979;197:319-22.<br />

4. Glaucoma Laser Trial Research Group. Acute effects of argon laser trabeculoplasty on intraocular pressure.<br />

Arch Ophthalmol 1989;107:1135-1142.<br />

5. Glaucoma Laser Trial Research Group. The Glaucoma Laser Trial, II: Results of argon laser trabeculoplasty<br />

versus topical medicines. <strong>Ophthalmology</strong> 1990;97:1403-1413.<br />

6. Glaucoma Laser Trial Research Group. The Glaucoma Laser Trial,VI: Treatment group differences in<br />

visual field changes. Am J Ophthalmol 1995;120:10-22.<br />

7. Glaucoma Laser Trial Research Group, VII: The glaucoma laser trial (GLT) and glaucoma laser trial follow-up<br />

study results. Am J Ophthalmol 1995;120:718-731.<br />

8. Brubaker RF, Liesegang TJ. Effect of trabecular photocoagulation on the aqueous humor dynamics of the<br />

human eye. Am J Ophthalmol 1983;96:139-47.<br />

9. Greenridge KC, et al. Acute intraocular pressure elevation after argon laser trabeculoplasty and iridectomy:<br />

a clinicopathologic study. Ophthalmic Surg 1984;15:105.<br />

10. Yablonski ME, Cook DJ, Gray BS. A fluorophotometric study of the effect of argon laser trabeculoplasty<br />

on aqueous humor dynamics. Am J Ophthalmol 1985;99:579-82.<br />

11. Bylsma SB, Samples JR, Acott TS, Van Buskirk EM. Trabecular cell division after argon laser trabeculoplasty.<br />

Archives of <strong>Ophthalmology</strong> 1988;106:544-547.<br />

12. Van Buskirk EM. Pathophysiology of laser trabeculoplasty. Surv <strong>Ophthalmology</strong> 1989;33:264-272.<br />

13. Alvarado JA, Murphy CG. Outflow obstruction in pigmentary and primary open angle glaucoma. Arch<br />

Ophthalmol 1992;110:1769-1778.<br />

14. Rodriguez MM, Spaeth GL, Donohoo P. Electron microscopy of argon laser therapy in phakic open<br />

angle glaucoma. <strong>Ophthalmology</strong> 1984;89:198-209.<br />

15. Van Buskirk EM, Pond V, Rosenquist RC, Acott TS. Argon laser trabeculoplasty – studies of mechanism<br />

of action. <strong>Ophthalmology</strong> 1984;91:1005-10.<br />

16. Melamed S, Pei J, Epstein DL. Short-term effect of argon laser trabeculoplasty in monkeys. Arch<br />

Ophthalmol 1985;103:1546-52.<br />

17. Brown SVL, Thomas JV, Simmons RJ. Laser trabeculoplasty re-treatment. Am J Ophthalmol 1985;99:8-10.<br />

18. Richter CU, Bradford JS, Bellows AR, Hutchinson BT, Jacobson LP. Retreatment with argon laser traculoplasty.<br />

<strong>Ophthalmology</strong> 1987;94:1085-1089.<br />

19. Starita RJ, Fellman RL, Spaeth GL, Poryzees E. The effect of repeating full-circumference argon laser<br />

trabeculoplasty. Ophthalmic Surg 1984;15:41-3.<br />

20. Jorizzo PA, Samples JR, Van Buskirk EM. The effect of repeat argon laser trabeculoplasty. Am J<br />

Ophthalmol 1988;106:682-5.<br />

21. Grayson DK, Camras CB, Podos SM, Lustgarten JS. Long-term reduction of intraocular pressure after<br />

repeat argon laser trabeculoplasty. Am J Ophthalmol 1988;106:312-21.<br />

22. Accott TS, Kingsley PD, Samples JR, Van Buskirk EM. Human trabecular meshwork organ culture:<br />

morphology and glycosaminoglycan synthesis. Invest Ophthalmol Vis Sci 1988;29:90-100.<br />

23. Anderson RR, Parrish JA. Microvasculature can be selectively damaged using dye lasers: a basic theory<br />

and experimental evidence in human skin. Lasers Surg Med 1981;1:263-76.<br />

25


24. Anderson RR, Parrish JA. Selective photothermolysis: precise microsurgery by selective absorption of<br />

pulsed radiation. Science 1983;220:524-7.<br />

25. Nakagawa H, Tan OT, Parrish JA. Ultrastructural changes in human skin after exposure to a pulsed<br />

laser. J Invest Dermatol 1985;84:396-400.<br />

26. Anderson RR, Margolis RJ, Watenabe S, Flotte T, Hruza GJ, Dover JS. Selective photothermolysis of<br />

cutaneous pigmentation by Q-switched Nd:YAG laser pulses at 1064, 532, and 355 nm. J Invest Dermatol<br />

1989;93:28-32.<br />

27. Diette KM, Bronstein BR, Parrish JA. Histologic comparison of argon and tunable dye lasers in treatment<br />

of tattoos. J Invest Dermatol 1985;85:368-73.<br />

28. Latina MA, Park C. Selective targeting of trabecular meshwork cells: In vitro studies of pulsed and CW<br />

laser interactions. Exp Eye Res 1995;60:359-372.<br />

29. Acott TS, Samples JR, Bradley JM, Bacon DR, Bylsma SS, Van Buskirk EM. Trabecular repopulation by<br />

anterior trabecular meshwork cells after laser trabeculoplasty. Am J Ophthalmol 1989;15:107.<br />

30. Bradley JM, Vranka J, Colvis CM, Conger DM, Alexander JP, Fisk AS, Samples JR, Acott TS. Effect of<br />

matrix metalloproteinases activity on outflow in perfused human organ culture. Invest Ophthalmol Vis Sci<br />

1998;39:2649-2658.<br />

31. Alexander JP, Samples JR, Acott TS. Growth factor and cytokine modulation of trabecular meshwork<br />

matrix metalloproteinase and TIMP expression. Curr Eye Res 1998;17:276-285.<br />

32. Bradley JM, Anderssohn AM, Colvis CM, Parshley DE, Zhu XH, Ruddat MS, Samples JR, Acott TS.<br />

Mediation of laser trabeculoplasty-induced matrix metalloproteinase expression by IL-1 beta and TNF<br />

alpha. Invest Ophthalmol Vis Sci 2000;41:422-430.<br />

33. Alvarado JA. Data on file, Coherent Medical. November, 2000.<br />

34. Noecker RJ, Kramer TR. Comparison of the acute morphologic changes after selective laser trabeculoplasty<br />

and argon laser trabeculoplasty in human eye bank eyes. In press, 2000.<br />

35. Latina MA. Data on file, Coherent Medical. May, 1996.<br />

36. Feder RS, Dueker DK. Can macrophages cause obstruction to aqueous outflow in rabbits? International<br />

<strong>Ophthalmology</strong> 1984;7:87-93.<br />

37. Parrish JA, Deutsch TF. Laser photomedicine. IEEE J Quantum Electronics 1984;12:1386-96.<br />

38. Damji KF, Shah KC, Rock WJ, Bains HS, Hodge WG. Selective Laser Trabeculoplasty v argon laser trabeculoplasty:<br />

a prospective randomised clinical trial. British Journal of Ophthalmol 1999;83:718-22.<br />

39. Kano K, Kuwayama Y, Mizoue S, Ito N. Clinical results of selective laser trabeculoplasty. Nippon Ganka<br />

Gakkai Zasshi 1999;103:612-616.<br />

40. Kim YJ, Moon CS. One-year follow-up of laser trabeculoplasty using q-switched frequency-doubled<br />

Nd:YAG laser of 523 nm wavelength. Ophthalmic Surgery and Lasers 2000;31:394-399.<br />

41. Lanzetta P, Menchini U, Virgili G. Immediate intraocular pressure response to selective laser trabeculoplasty.<br />

British Journal of <strong>Ophthalmology</strong> 1999;83:29-32.<br />

42. Latina MA, Sibayan SA, Shin DH, Noecker RJ, Marcellino G. Q-switched 532-nm Nd:YAG laser trabeculoplasty<br />

(Selective Laser Trabeculoplasty). <strong>Ophthalmology</strong> 1998;105:2082-2090.<br />

43. Hoskins HD, Hetherington J, Minckler DS, Lieberman MF, Shaffer RN. Complications of laser trabeculoplasty.<br />

<strong>Ophthalmology</strong> 1983;90:796-799.<br />

44. Krupin T, Kolker AE, Kass MA, Becker B. Intraocular pressure the day of argon laser trabeculoplasty in<br />

primary open-angle glaucoma. <strong>Ophthalmology</strong> 1984;91:361-365.<br />

45. Robin AL, Pollack IP, House B, Enger C. Effects of ALO 2145 on intraocular pressure following argon<br />

laser trabeculoplasty. Arch Ophthalmol 1987;105:646-650.<br />

26<br />

46. Weinreb R, Ruderman J, Juster R, Zweig K. Immediate intraocular pressure response to argon laser trabeculoplasty.<br />

Am J Ophthalmol 1983;95:279-286.


Global Headquarters<br />

Coherent Medical Coherent Japan, Inc. Coherent GmbH Coherent (UK) Ltd.<br />

2400 Condensa Street Tokyo, Japan Dieburg, Germany Ely, UK<br />

Santa Clara, CA 95051 USA TEL: (81) 3 5635 8600 TEL: (49) 6071 968 110 TEL: (44) 13 53 658824<br />

TEL: (408) 764-3000 FAX:(81) 3 5635 8601 FAX: (49) 6071 968 400 FAX: (44) 13 53 659106<br />

(800) 635-1313<br />

FAX: (408) 764-3999<br />

(800) 505-1133<br />

Coherent, Inc. Beijing Coherent (HK) Ltd. Coherent Scandinavia Coherent Médical S.A.R.L.<br />

Beijing, China Kowloon, Hong Kong Kristianstad, Sweden Saclay, France<br />

TEL: (86) 10 6510 2620 TEL: (852) 2174 2800 TEL: (46) 44 21 21 26 TEL: (33) 1 69 85 53 52<br />

FAX: (86) 10 6510 2621 FAX:(852) 2722 5151 FAX: (46) 44 21 21 79 FAX: (33) 1 69 85 35 93<br />

Internet Address<br />

www.CoherentInc.com<br />

email: cmg.web@cohr.com<br />

Acknowledgements: Authored by Karen Baker, Senior Manager, Regulatory Affairs,<br />

Coherent Medical; George Marcellino, Ph.D.; Judy Gordon, D.V.M.<br />

Reviewed by Jorge Alvarado, M.D., University of California at<br />

San Francisco and Mark A. Latina, Tufts University School of Medicine.<br />

Coherent, its logo and Selecta are registered trademarks of Coherent Inc.<br />

Coherent © 2001. Specifications are subject to change without notice.<br />

Printed in U.S.A. M10524012.5M<br />

93/42/EEC<br />

0459<br />

Nd:YAG LASER: Laser Class 3B/IIIb<br />

MAX OUTPUT: 532 nm, 5 mJ, 3 ns pulse<br />

DIODE LASER: Laser Class 2/II<br />

MAX OUTPUT: 635 nm,

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!