retinal laser therapy biophysical basis

Upload: andrian-hanciu

Post on 14-Apr-2018

237 views

Category:

Documents


1 download

TRANSCRIPT

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    1/15

    Chapter

    39

    INTRODUCTION

    Historically, a number o light sources have been utilized orretinal phototherapy, including the sun, various ash lamps, and

    lasers. The sun is capable o producing a retinal burn either

    accidentally (e.g., in the case o solar eclipse retinopathy) or onpurpose, as demonstrated frst by Meyer-Schwickerath.1

    However, its dependence on weather conditions, its constantmotion in the sky, and relatively large angular size (0.52) make

    it an impractical method or intraocular therapy.Arc lamps are very bright sources o light used in many thera-

    peutic and imaging applications. In such lamps, a high-voltagedischarge initially ionizes gas between an anode and cathode,

    creating an electric arc a high-current-density discharge in thelamp. The ions emit light at specifc wavelengths, and the spec-

    trum o the plasma emission depends on the types o atoms

    involved, their temperatures, and gas pressure. Thus the spec-trum o an arc lamp may have a distinct signature. The xenon arc

    lamp was the frst to become widely used or retinal photocoagu-lation because o its strong visible and near-inrared emission,

    convenience, and low price. However, because o its large sizeand tendency to produce intense retinal burns, it was replaced in

    clinical practice by laser-based systems in the early 1970s.

    2

    Lasers became the preerred light source or retinal photoco-

    agulation due their narrow spectrum, wide selection o wave-lengths, excellent collimation (directionality), high brightness,

    and variable pulse duration. The directionality o the laser makes

    it easy to manipulate the beam optically beore its introductioninto the eye, and to ocus it into very small spots. Its monochro-

    maticity makes it possible to choose a wavelength or selectiveabsorption in specifc tissues o the eye. Adjustable pulse dura-

    tion allows limiting the thermal diusion to small distances, thusproducing very precise and selective interactions with minimal

    collateral damage.The most widespread medical applications o lasers in medi-

    cine have been in ophthalmology. Since the introduction o the

    ruby laser more than three decades ago, ophthalmic laser appli-cations have experienced rapid growth with the use o argon,

    krypton, argon-pumped dye, Nd:YAG, diode, Er:YAG, excimer,and Ti:sapphire lasers. Lasers have been applied to a wide

    variety o slit-lamp-based retinal therapies, as well as to vitreo-retinal surgery, glaucoma, lens capsule opacifcation, and rerac-

    tive surgery. These applications are based on dierentmechanisms o lasertissue interactions including photothermal,

    photodisruptive, and photochemical interactions. The mostcommon vitreoretinal application is retinal photocoagulation.

    Additionally, a number o novel therapies have recently been

    Retinal Laser Therapy: Biophysical Basisand ApplicationsDaniel Palanker, Mark S. Blumenkranz

    Section 4 Translational Basic Science

    introduced and are under active evaluation, including select

    retinal pigment epithelium (RPE) treatment and subleththermal therapy.

    In the ollowing sections we will describe the underlying pr

    ciples o lasertissue interactions and the types o lasers avable and appropriate or various vitreoretinal applications.

    Optical properties of the eyeThe relaxed eye has an approximate optical power o 60 D (i

    its ocal length is 16.7 mm in air), with the corneal power beiabout 40 D, or two-thirds o the total power.3 Due to ordearrangement o collagen fbrils in the cornea it is highly transp

    ent, with transmission above 95% in the spectral range 400900 nm.4 The reractive index o the cornea is n 1.376

    0.0005.4 The amount o light reaching the retina is regulated

    the pupil size, which can vary between 1.5 and 8 mm. The anrior chamber o the eye, which is located between the cornea a

    lens capsule, is flled with a clear liquid the aqueous humwhich has a reractive index n 1.3335. The crystalline lens

    the eye, located behind the iris, is composed o specialized crtallin proteins with reractive index o n= 1.401.42. The lens

    about 4 mm in thickness and 10 mm in diameter and is enclos

    in a tough, thin (515 m), transparent collagenous capsule.the relaxed eye the lens has a power o about 20 D, while in tully accommodated state it can temporarily increase to 33

    The vitreous humor, a transparent jelly-like substance flling t

    large cavity posterior to the lens, and anterior to the retina, ha reractive index n 1.335.4

    Light entering the eye can be reected, scattered, transmittor absorbed. Reected or scattered light contains inormati

    that can be used or noninvasive diagnostic purposes. Tabsorption characteristics o ocular tissues are determined by t

    chromophores resident within the tissue. In the visible partthe spectrum (400800 nm) these chromophores include:

    melanin located in the retinal and iris pigmented epithe

    choroid, uvea, and trabecular meshwork; (2) hemoglobin, loca

    in the red blood cells; (3) macular xanthophyl, which is locatin the plexiorm layers o the retina, especially near and in macula; (4) rhodopsin and cone photopigments which a

    located in the photoreceptors; and (5) lipouscin, located primily in the RPE layer. These pigments are o major importance

    absorption o visible light in the retina rom both physioloand pathologic standpoints. The absorption spectra o these p

    ments, as well as water and proteins, are illustrated in Figu39.1. In the mid-inrared part o the spectrum (315 m)

    major absorber is water, while in ultraviolet (below 250 n

    protein absorption is dominant.

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    2/15

    BASICS OF LASERS

    The term laser stands or light amplifcation by stimulatedemission o radiation. A beam o light is composed o individual

    packets o energy that are called quanta or photons. Each othese photons has a particular energy and direction o travel. The

    energy o a quantum o light is proportional to its requency, i.e.,

    it is reciprocal o its wavelength. In the presence o a properlyprepared laser material, it is possible or a quantum o light to

    trigger the release o other quanta with the same wavelength and

    direction o travel. This phenomenon is called stimulated emis-sion, and it is an essential element in lasing. In thermal equilib-rium the energy levels o atoms and molecules are populated

    according to the Boltzmann distribution, in which upper levelsare always less populated than the lower levels. Stimulated

    emission requires an inverted population o energy levels, suchthat the upper levels are more populated than the lower ones.

    As a result, lasing can occur only when material is not in thermal

    equilibrium. The nonequilibrium state is created in the lasingmaterial by an excitation source or pump.

    Generally, a laser is composed o three basic components:(1) a material that can store energy to be released by stimulated

    emission; (2) a means o replenishing the energy stored in the

    lasing material; and (3) some method o retaining a raction othe light emitted by the lasing material to stimulate urther emis-sion. Figure 39.2 schematically illustrates a general confguration

    o a laser. An energy source is used to introduce energy into thelasing material. This energy is stored as atomic or molecular

    excitation waiting to be released by stimulated emission. Laser

    light that has already been emitted by the lasing material circu-lates between the two mirrors on either end o the laser cavity,

    with a raction o the light escaping through one mirror to ormthe laser beam. The trapped light stimulates emission o new

    quanta o light rom the laser material with the same wavelengthand direction as the original quanta. In this way, a laser produces

    a beam o light between the two mirrors in which all o thequanta move in phase with one another. This property o light

    is called coherence.Coherence is related to synchronization o light in time, or

    along the laser beam. The duration o the synchronized emission

    rom the laser multiplied by the speed o light is called the coher-ence length o the laser emission. This is the distance along

    Fig. 39.1 (A) Absorption coefcients or major chromophores in aspectral range o 0.210 m. (B) Absorption coefcients o the majorocular chromophores in the visible part o the spectrum: 400900 nm.Spectral locations o some o the popular laser lines in this range areindicated above the plots.

    10000

    diode

    805 nm

    Krypton

    647 nm

    Krypton

    568 nm

    Argon

    514 nm

    Argon

    488 nm

    Wavelength, nm

    1000

    Hb

    Hb02

    Melanin

    Xantrophyll

    Water

    Proteins

    Hb

    Hb02

    Melanin

    1E-4

    1E-3

    0.01

    0.1

    1

    10

    100

    1000

    10000

    1400 500 600 700 800 900

    Wavelength, nm

    10

    100

    1000

    Excimer

    193 nm

    Nd: YAG-2nd

    532 nm

    Nd: YAG

    1064 nm

    Er: YAG

    2.94 m

    CO2

    10.6 m

    Absorptioncoefficient,a

    (cm

    1)

    Absorptioncoefficient,a

    (cm

    1)

    500200

    A

    B

    Fig. 39.2 Laser typically consists o theenergy source (pump), the lasing medium,and the optical cavity with a partiallytransparent ront mirror.

    Mirror

    100%

    Mirror

    90%

    Pumping energy

    Output beamLasting medium

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    3/15

    748

    Section

    4

    TranslationalBasicScience

    BasicS

    cience

    and

    Translation

    to

    Therapy

    Fig. 39.3 Laser beam o diameter Docuswith a lens o ocal length fproduces a wawith diameter dand a ocal depth F(seeequations in the text).

    D

    F

    f

    d

    Fig. 39.4 Optical fber typically consists o core, cladding, and a jacket (buer coatingLight launched into the fber with itsacceptance cone ac is trapped within a codue to the total internal reection at thecorecladding interace.

    Jacket (buffer coating)

    Cladding

    CoreAcceptance

    cone ac

    which the photons are coherent, or moving in step. To remain

    in phase with one another, these quanta must have approxi-mately the same wavelength. Thus temporal coherence is related

    to the monochromaticity (or spectral width) o the light emittedrom the laser: the broader the spectrum, the shorter the temp-

    oral coherence. A laser may produce one or several discrete

    spectral lines in the inrared, visible, or ultraviolet domains, incontrast to conventional light sources (incandescent or arc

    lamps) which typically produce noncoherent polychromatic

    (broadband or white) light.Collimation (directionality) o the emitted beam is governedby the mirror confguration o the laser cavity. In its simplest

    orm, a cavity consists o two mirrors arranged such that lightbounces back and orth, each time passing through the gain

    medium. One o the two mirrors, the output coupler, is par-tially transparent, allowing the output beam to exit through

    it (Fig. 39.2). The reection coefcient o the output coupler

    determines how many times photons are reected back tocirculate inside the cavity beore exiting it. For example, with

    a reection coefcient o 0.99, the photon will bounce, onaverage, 99 times beore exiting the cavity. The structure o

    the laser cavity determines directionality (collimation) o thelaser beam, which determines its ability to be ocused into a

    small spot.The lasing medium can be a gas, liquid, or solid. Lasers can

    be pumped by continuous discharge lamps and by pulsed ashlamps, by electric discharges in the laser medium, by chemical

    reactions, by an electron beam, by direct conversion o electric

    current into photons in semiconductors, or by light rom otherlasers. Laser pulse durations can vary rom emtoseconds to

    infnity. Pulsing techniques used or dierent ranges o pulsedurations include electronic shutters (down to 1 ms), pulsed

    ash lamps (typically down to a ew s), Q-switching (down toa ew ns), or mode-locking (down to s).

    Laser beam delivery to tissueLaser beams are typically very well collimated. Diracti

    causes light waves to spread transversely as they propagate, a

    it is thereore impossible to have a perectly collimated beaThe diraction-limited divergence angle o a gaussian beam w

    diameter D and wavelength is =

    4

    D. As an example,

    an argon laser emitting a 1-mm-wide beam at 515 nm wav

    length, the divergence (hal-angle ) is about 0.66 mrad, i.e., tbeam spreads by 1.3 mm over a distance o 1 meter.

    Using a lens or a concave mirror with ocal length f, a la

    beam can be ocused to a spot with a diameter df

    D=

    4

    . T

    depth o the ocal region is Ff

    D=

    82

    2

    (Fig. 39.3). With

    f= 25 mm lens the same argon laser beam can be ocused

    a spot o 16 m in diameter, having a ocal depth o 820 It must be emphasized, though, that exact defnition o

    spot size depends on the beam profle, which varies in vario

    confgurations o laser cavities. For therapeutic laser photoagulation such tight ocusing is usually not required, and la

    spots typically vary in diameter between 50 and 500 m

    various applications.As an alternative to a ree-propagating beam, laser light c

    be transported via optical fbers. An optical fber, schematica

    shown in Figure 39.4, typically consists o a core, cladding, ajacket. Light is trapped within the core due to total inter

    reection at the interace o core with cladding. To satisy contions o total internal reection, the incidence angle o light

    the corecladding interace should not exceed the critical an

    o total internal reection: sincr=ncore/nclad, where ncore and nare reractive indices o the core and cladding, respectively.

    satisy these criteria or total internal reection, the light shou

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    4/15

    be launched within a so-called acceptance cone, and the sine o

    its hal-angle ac is defned as the numerical aperture (NA) othe fber: NA n nac core clad= = sin

    2 2 . Typically the NA is within

    a range o 0.10.2. Optical fbers are oten used or delivery o

    laser light to slit-lamp-based systems (Fig. 39.5), and to intraocu-lar surgical probes (Fig. 39.6).

    ABERRATIONSWith nonperect ocusing optics, the ocal spot size o the laserbeam is limited not only by diraction, but also by aberrations.

    Measurements o optical aberrations in the human eye demon-strate5,6 that or pupillary dilation o up to 3 mm in diameter,

    an average emmetropic human eye is optically well corrected,

    and the ocal spot is close to the diraction limit. However, orpupils greater than 3 mm in diameter, central aberrations

    increase, resulting in increases in the ocal spot size. Peripheralfeld aberrations lead to rapidly increasing blur o the image

    with angle o visual feld, strongly limiting the ocusing

    Fig. 39.6 Intraocular hand pieces and optical fber or vitreoretinaldissection with the Er:YAG laser.

    Table 39.1 Ocular contact lenses and their magnifcationsin a human eye

    LensImagemagnifcation

    Laser beammagnifcation

    Ocular Mainster Standard 0.95 1.05

    Ocular Fundus Laser 0.93 1.08

    Ocular Mainster Wide Field 0.67 1.50

    Ocular Mainster Ultra Mag 0.53 1.90

    Ocular Mainster 165 0.51 1.96

    Ocular Three Mirror Universal 0.93 1.08

    Volk G-3 Gonio Fundus 1.06 0.94

    Volk Area Centralis 1.06 0.94

    Volk Trans Equator 0.69 1.44

    Volk SuperQuad 160 0.5 2.00

    Volk QuadrAspheric 0.51 1.97

    Volk High Resolution WideField

    0.5 2.00

    Rodenstock Panundoscope 0.67 1.50

    Fig. 39.5 Laser photocoagulation on a slit-lamp system. 1, Optical fberand electronic cable connecting laser with a slit-lamp system; 2, opticalcoupler projecting the beam exiting rom the fber on to the retina;3, contact lens.

    capability o laser in the periphery o the retina.6 In retinal

    photocoagulation, a at contact lens is typically used to reducethe optical power o the ront surace o the cornea. I the lens

    is used properly it aids greatly in controlling peripheral aber-rations during photocoagulation. Other methods include using

    an aspheric lens to control optical aberrations in the peripheryduring photocoagulation. The use o such lenses has many

    advantages, in particular or providing wide-feld viewing,

    although aberrations are difcult to correct over the totality ofelds o interest and additional reections may be introduced

    by the lens suraces.

    CONTACT LENSES

    Currently, retinal laser photocoagulation relies heavily on the

    use o contact lenses. A number o contact lenses have beendeveloped or this purpose, and the most common types are

    listed in Table 39.1. The universal (Goldmann) three-mirrorcontact lens provides a at ront surace that nearly cancels

    the positive reractive power o the ront surace o the cornea.Mirrors at 5, 67, and 73 aid in visualization and photoco-agulation o the periphery and anterior segment. To obtain the

    most reproducible results in photocoagulation the operatorshould hold the contact lens so that the at surace is within

    5 o perpendicular to the laser beam (Fig. 39.5). The use omirrors in contact lenses helps the operator keep the laser beam

    properly aligned to the lens while photocoagulating over alarge feld.

    Another useul photocoagulation lens is the inverted image

    lens system, typifed by the Rodenstock, Quadraspheric, and

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    5/15

    750

    Section

    4

    TranslationalBasicScience

    BasicS

    cience

    and

    Translation

    to

    Therapy

    Mainster photocoagulation lenses. These lenses contain a lens

    element in contact with the corneal surace and another posi-tive lens element at a fxed distance rom the cornea. These

    systems magniy the spot size on the retina, while increasingthe feld o view, requiring the operator to adjust the power

    accordingly. Magnifcation actors o the most common contact

    lenses are listed in Table 39.1. It is important to keep in mindthat magnifcation o the retinal image demagnifes the beam

    size on the retina by the same amount: the higher the

    magnifcation o the retina, the smaller the laser spot onthe retina.

    INTERACTIONS OF LIGHT WITH TISSUE

    In linear interactions the irradiance (or power uence) I(z) (W/cm2) o the beam propagating inside tissue decreases exponen-

    tially with depth (Beers law) due to absorption and scatteringo light: I(z) = (1 ks)I0exp(z), where I0 is the light intensity at

    the surace o tissue (z= 0), ks is the specular reection coefcient

    at the tissue surace, =a+s is the attenuation coefcient,combined o absorption and scattering components. The pene-

    tration depth () o the beam into tissue is defned as a depth atwhich light intensity is reduced by a actor o e: = 1/. Reec-

    tion o light rom ocular tissue at normal incidence typically doesnot exceed 2%. As shown in Figure 39.1, absorption o light by

    various chromophores in the eye strongly varies with wave-length. Scattering o light is also a very strong unction o the

    wavelength: scattering on subwavelength inhomogeneities oreractive index in ocular media (e.g., collagen fbrils) is recipro-

    cal to the ourth power o the wavelength: s ~ 4 (Rayleigh

    scattering). For example, the scattering coefcient or light at1064 nm is 16 times lower than that at 532 nm. Scattering rom

    structures larger than the wavelength (e.g., cellular organelles)is described by Mie theory and has more complex wavelength

    dependence and spatial distribution. Scaling o the Mie scatter-ing coefcient with wavelength in various tissues can be approx-

    imated as s ~ b

    , with b 0.52.7,8

    Photochemical interactionsPhotochemical interactions are based upon nonthermallight-induced chemical reactions. The best-known natural pho-

    tochemical reactions are photosynthesis in plants and photo-

    transduction in photoreceptors. Therapeutic photochemicalinteractions used in photodynamic therapy (PDT) take place at

    very low power densities (typically

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    6/15

    Photothermal interactionsTemperature is the governing parameter in all thermal laser

    tissue interactions. Depending on the duration and peak valueo the temperature, dierent tissue eects, including necrosis,

    coagulation, vaporization, carbonization, and melting, mayoccur.

    Heat generation in tissue is determined by the laser parame-ters and optical tissue properties irradiance, exposure time,

    and the absorption coefcient, which is a unction o the laser

    wavelength. Heat transport is characterized by heat conductivityand heat capacity. Heat eects depend on the type o tissue and

    temperature history (values and durations).Absorption o light in tissue leads to heating. I heat diusion

    is not taken into account, then at a constant beam intensity the

    temperature rise is linear with time: T z tI z t

    c

    a,( )

    ( ) =

    , where

    is tissue density, and c is its heat capacity (c = 4.2 J/g/K,

    = 1 g/cm3 or water). To assess whether heat diusion playsa signifcant role during the laser pulse, one should compare

    pulse duration with a characteristic time it takes or heat tospread by the distance equal to the zone o initial heat deposi-

    tion in tissue. For the heated zone (laser penetration depth) o

    length L, the heat diusion time is: = L2

    /4, where isthermal diusivity (= 1.4103 cm2/s or water). For example,

    or L= 1 m in water the characteristic heat diusion time ist= 1.7 s, while or L= 1 mm the diusion time t= 1.7 s. I

    the laser pulse duration is comparable or longer than the char-acteristic diusion time across the light absorption zone, then

    proper estimation o the peak temperature in tissue shouldtake heat diusion into account.

    Sublethal thermotherapyThere is a growing body o clinical evidence that diabetic macular

    edema can be successully treated by pulsed near-inrared diodelaser (810 nm) without producing visible lesions.1721 Using

    300-ms bursts o submillisecond micropulses, laser energy is

    applied with no visible lesions and no uorescein leakage, as

    treatment o classic and occult CNV. Verteporfn has a very

    broad absorption spectrum, but only the ar-red peak at688691 nm is typically utilized in clinical practice (Fig. 39.8).

    This is because o the lower sensitivity o retina to ar-red lightand its superior penetration into the choroid.14

    In PDT treatment with verteporfn activation by laser is typi-

    cally perormed 1520 minutes ater the intravenous injection othe dye. A beam o red laser light (689 nm diode laser) is applied

    to the retina via a slit-lamp delivery system, irradiating a spot

    chosen to exceed the dimension o the neovascularization mini-mally, with light intensity o 600 mW/cm2, or 83 seconds,resulting in a total radiant exposure o 50 J/cm2.15,16 Closure o

    abnormal (leaking) blood vessels occurs or approximately 612weeks in most patients. Reperusion is common and multiple

    treatments are oten required. Figures 39.9A (pretreatment) and39.9B (1 week posttreatment) showing uorescein angiography

    images demonstrate closure o a suboveal CNV membrane ater

    PDT.

    Fig. 39.9 (A) Fluorescein angiogram o a patient with predominantly occult suboveal choroidal neovascular membrane in let eye. (B) Same eye1 week ollowing photodynamic therapy with verteporfn and intravitreal triamcinolone injection. Note absence o hyperuorescence in the area oprevious neovascularization and subtle darkening o choroid corresponding to area o photodynamic closure o the membrane.

    BA

    Fig. 39.8 Absorption spectrum o verteporfn. Arrow indicates the

    689-nm peak typically used or photodynamic therapy.

    Wavelength, nm

    300 400 500 600 700 8000

    10

    20

    30

    40

    50

    60

    70

    80

    90

    Molarextinctioncoefficient[nM

    -1cm-1]

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    7/15

    752

    Section

    4

    TranslationalBasicScience

    BasicS

    cience

    and

    Translation

    to

    Therapy

    observed acutely and in subsequent clinical exams. However,

    application o higher power can lead to signifcant heat accumu-lation and result in damage to RPE and photoreceptors. 17,22

    Avoidance o acute laser-induced retinal damage permits apply-ing this treatment conuently on the target retina with a large

    number o small (125 m), densely placed laser spots.19,23 The

    absence o damage allows this therapy to be repeated as neces-sary, and makes this technique potentially appealing in applica-

    tions proximal to the ovea. However, due to the lack o reliable

    dosimetry and observable measures or such subvisibletreatment, this technique is difcult to optimize, leading topotentially broad variability in the outcomes.

    NecrosisTemperature rises induce conormational changes in various

    proteins, which denature at characteristic rates specifc toprotein species. These thermal processes, which may eventually

    lead to cell necrosis, depend on both the temperature and dura-tion o the hyperthermia. Thermal cellular damage in a milli-

    second regime can be approximated using the Arrheniusmodel.24,25 It assumes a rate o decline in the concentration o a

    critical molecular component or cellular metabolism D(t) with

    temperature T(t):

    dD t D t AE

    R T tdt( ) ( ) exp

    ( )=

    *(1)

    where E* and A are the activation energy and rate constant

    setting parameters to the process, and R is the gas constant,8.3 J/(K mol). Tissue damage, i.e., decrease in critical molecular

    component D(), relative to its initial value D0 over the pulselength is encapsulated in the Arrhenius integral :

    ( ) ln( )

    exp( )

    =

    =

    D

    DA

    E

    R T tdt

    0 0

    *(2)

    The model assumes that irreversible tissue damage takes placewhen concentration o the critical molecular component drops

    below some threshold value. Conventionally, this threshold cor-responds to a reduction in concentration by a actor o e, or an

    Arrhenius integral o unity. Thus when = 1, the nondenatured

    raction o proteins is 1/e 37%, or in other words, 63% o pro-teins have been damaged.

    Measurements o the RPE damage at various irradiation con-ditions yields the ollowing average values25: E* = 340 kJ/mol,

    A= 1.6 1055. It is important to keep in mind, though, that accu-rate estimation o cell survival under thermal stress is much

    more complex than just assessment o the denaturation rate oone type o protein or another. For example: (1) there are mul-

    tiple types o proteins in cells and they denature at dierent

    rates; (2) dierent proteins have dierent importance or cellularsurvival; (3) cellular repair mechanisms cannot be ignored at

    long exposures. Thereore the single values o the reaction rateA and activation energy E most likely represent characteristics

    o the weak link in cellular metabolism, most susceptible tothermal damage.

    Figure 39.10 shows an example o the temperature rise intissue or a hypothetical square pulse o heating, which is su-

    fcient or cell death, according to the Arrhenius model param-eters listed above. Cells exposed to temperatures above the

    threshold curve are coagulated and the tissue becomes necrotic.

    Fig. 39.10 Solid line depicts Arrhenius approximation o the cellulardamage threshold as a unction o duration o a hypothetical squarepulse o heating. Dashed line indicates deviation o the damagethreshold rom the Arrhenius model at long exposures.

    Sublethal thermal stress

    Irreversible damage

    T

    emperature,

    C

    40

    45

    50

    55

    60

    65

    70

    75

    80

    1.E-04 1.E-03 1.E-02 1.E-01 1.E-00

    Time, s

    1.E+0

    For example, at 1 second long exposure T 50C, while at 10 it requires 67C or a lethal thermal damage. This curve

    approximate, and the exact values depend on the shape o tactual pulse o temperature, type o cells, and tissues involve

    The Arrhenius model ails to predict correct threshold tempetures at exposures longer than approximately 1 second, sin

    cellular repair mechanisms should be taken into account at loexposures.

    Transpupillary thermotherapyThe possibility o localized tissue coagulation and necrosis or

    the basis o the tumor treatment technique called laser-inducinterstitial thermotherapy (LITT) or transpupillary thermoth

    apy (TTT). It has been applied to tumors in retina, brain, prtate, and liver, and is considered a orm o minimally invas

    surgery. The concept o LITT is to apply a laser beam to tiss

    in such a way that the target tissue is heated or a prolongperiod o time (about 1 minute) to temperatures above t

    threshold o necrosis (on the order o 60C). To achieve depenetration into tissue continuous wave lasers in the ne

    inrared region (8001064 nm) are typically employed. TTT the treatment o intraocular tumors26 typically requires exposu

    times o 1 minute and irradiances varying rom 5 to 12 W/cmThe use o TTT has been tested in the treatment o CNV

    AMD.15,27,28 Proponents o this approach have hypothesized

    selective eect on the heating o actively dividing cells in newormed blood vessels due to their higher susceptibility to therm

    injury than nondividing cells in normal tissue. The estimatretinal temperature elevation with TTT at standard settin

    (810 nm, 800 mW, 60 seconds, 3.0 mm spot size) is appromately 10C.29,30 The mechanism o treatment o CNV by T

    may occur through vascular thrombosis, apoptosis, or thermal inhibition o angiogenesis.31

    PhotocoagulationRetinal photocoagulation typically involves the application

    laser pulses with durations ranging rom 10 to 200 ms, and trsient hyperthermia by tens o degrees above body temperatu

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    8/15

    Fig. 39.11 Histology o the rabbit retina at1 day (let column) and 4 months (rightcolumn) ater photocoagulation. Retinal spotsize is 330 m, power 175 mW. (A) Intenseretinal burn produced with 100 ms exposure.

    Yellow bar shows the lateral extent o thelesion. Note ull-thickness retinal injury,including the inner retinal layers. (B) Lightburn produced with 15 ms exposure.Photoreceptors are coagulated, while innerretina is well preserved. (C) Barely visiblelesion produced with 7 ms pulse. Rightcolumn shows corresponding retinal scarringat 4 months. Note complete closure o thedamage zone by shiting photoreceptors inthe barely visible lesion.

    A

    B

    C

    1 day100 m

    4 months

    Various lasers have been used in the past (ruby (694 nm), argon

    (488, 514 nm), krypton (647 nm)). Currently the most commonlasers in photocoagulation are requency-doubled Nd:YAG

    (532 nm), and yellow semiconductor laser (577 nm). The laserenergy is absorbed primarily by melanin in the RPE and choroid,

    and by hemoglobin in blood. At a 532-nm wavelength approxi-

    mately hal o the laser energy incident on the retina is absorbedin the RPE, and the rest in the choroid.25 The heat generated di-

    uses rom the RPE and choroid into the retina and causes coagu-

    lation o the photoreceptors and, sometimes, o the inner retina.During 100-ms applications, the heat diuses distances o up to200 m, thus smoothing the edge and extending the coagu-

    lated zone beyond the boundaries o the laser spot, termedthermal blooming. Heat diusion using shorter pulses and

    with smaller spot sizes can be limited to the photoreceptor layer,thereby avoiding the inner retinal damage.

    The let panel in Figure 39.11A demonstrates the acute eects

    o an intense burn in a rabbit retina produced by 100 ms laserapplications, including ull-thickness injury and early necrotic

    eatures 24 hours ater treatment. The let panel in Figure 39.11Bdemonstrates a light lesion produced by 15 ms pulses. Damaged

    photoreceptors are pyknotic, but the inner nuclear layer andganglion cell layer are very well preserved.

    Figure 39.12 illustrates the eect o laser power and pulseduration on the size o the coagulated zone in pigmented

    rabbits.32Table 39.2 lists the ratio o the lesion width to the retinalbeam size or lesions o various clinical grades in human patients,

    as measured by optical coherence tomography (OCT) within 1

    hour o treatment.33 As can be seen, lesion size increases relativeto the beam width with more intense lesions and longer pulses.

    The threshold power required or the creation o retinal lesions

    increases with shorter pulses, since higher temperatures arerequired or coagulation during shorter exposures. An example

    o the threshold powers or lesions o various grades is plottedin Figure 39.13 as a unction o pulse duration or a 132 m

    retinal laser spot in the rabbit. A relatively modest power

    increase is required to produce comparable lesion grades goingrom 100 to 10 ms, whereas a much steeper increase is seen or

    durations under 10 ms. For pulse durations o 20, 50, and 100 ms,

    all the grades (mild, moderate, intense, very intense, and rupture)could be created with appropriate choice o power settings. Atpulse durations below 10 ms, it became increasingly difcult to

    create intense lesions reproducibly without inadvertently rup-turing the retina. At 2 ms or less it was not possible to create a

    moderate lesion reproducibly without rupturing the retina. At1 ms, there was little or no dierence between the power required

    to create a mild retinal lesion or produce a rupture.

    The ratio o the threshold power required to produce a ruptureto that required to produce a mild lesion is defned as the thera-

    peutic window, and represents one means o quantiying therelative saety (dynamic range) o retinal photocoagulation. The

    larger this ratio, the greater the margin o saety to create avisible lesion without inadvertently inducing a retinal rupture.

    Figure 39.14 depicts the width o this therapeutic window as aunction o pulse duration or two dierent laser spot sizes. For

    a 132 m retinal laser beam size, as pulse duration decreasesrom 100 to 20 ms, the width o the therapeutic window declines

    rom 3.9 to 3.0. When pulse duration is urther decreased to

    10 ms, the therapeutic window decreases urther to 2.5, and itapproaches unity at a pulse durations o 1 ms. For a 330 m

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    9/15

    754

    Section

    4

    TranslationalBasicScience

    BasicS

    cience

    and

    Translation

    to

    Therapy

    Fig. 39.12 (A) Retinal lesions in the rabbit eye with variable power and duration o exposure. Retinal beam size is 132 m. (B) Lesion diameteas a unction o laser power and duration o exposure. Laser beam size on the retina is 132 m (indicated by dashed line and arrow).

    250 mW

    200 mW

    150 mW

    100 mW

    50 mW

    100908070605040

    Pulse duration, ms

    3020100

    500

    400

    300

    200

    100

    0

    Burndiameter,m

    BPulse duration, ms

    10 20 50 100

    Power

    A

    Table 39.2 Ratio o the lesion width to retinal beam size or various pulse durations and clinical grades, as measured by optical coherencetomography in human patients within 1 hour o application. Coagulation was perormed with Area Centralis lens (laser beam magnifcation0.94)

    Beam size Lesion clinical grade

    In air On retina Moderate Light Barely visible

    100 ms 20 ms 100 ms 20 ms 100 ms 20 ms

    100 m 94 m 3.81 0.98 2.50 0.30 2.08 0.24

    200 m 188 m 2.08 0.22 1.49 0.09 1.24 0.08 0.93 0.08

    400 m 376 m 1.39 0.08 1.15 0.07 1.19 0.11 0.99 0.09 0.99 0.08 0.74 0.12

    Fig. 39.13 Threshold power o retinal photocoagulation in rabbit eye,as a unction o pulse duration. Laser beam size on the retina is132 m. Clinical grades indicated by the colors: light, moderate,intense, very intense, and rupture.

    Power,mW

    Light

    Moderate

    Intense

    Very intense

    Rupture

    Pulse duration, ms

    1001010

    50

    100

    150

    200

    250

    300

    350

    400

    Fig. 39.14 Sae therapeutic window o retinal photocoagulation (ratioo the threshold o rupture to that o light coagulation) increases withpulse duration, and with a beam size on the retina (shown or 132 a330 m).

    5

    4

    3

    2

    1

    1 10 10

    132 um

    330 m

    Pulse duration, ms

    Safe

    Unsafe

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    10/15

    Patterns included square arrays with up to 5 5 spots, arcs

    with the number o concentric rows varying rom 1 to 3, andcircular patterns or photocoagulation o small holes and other

    lesions in the retinal periphery. Patterns or macular photoco-agulation included rings and arcs with an adjustable central

    exclusion zone o up to 2 mm in diameter to allow or laser

    application, reducing the risk o inadvertent damage to theoveal avascular zone.

    To deliver the whole pattern within the eye fxation time and

    avoid beam movement due to the ocular muscles, each exposurewas required to be shorter than in conventional photocoagula-tion: 1020 ms instead o 100200 ms, traditionally applied with

    single spot exposures. Reduced heat diusion into choroidduring shorter exposures also resulted in patients experiencing

    less pain.3941 Short pulse lesions appear smaller and lighter thanconventional burns produced with the same beam size, and

    thereore a larger number o them are required to treat the same

    total area.33

    An automatic laser delivery, guided by diagnostic imaging

    and stabilized using eye tracking, has been recently introducedin a Navilase system (OD-OS). This system includes retinal

    image acquisition, annotation o the images to create a detailedtreatment plan, and then automated delivery o the laser to the

    retina according to the treatment plan.

    Clinical indications: treatment of diabetic retinopathy

    Photocoagulation has proven sae and eective in the treatmento prolierative diabetic retinopathy. In this disorder the retina

    becomes ischemic and releases a variety o chemical messengers,most importantly, vascular endothelial growth actor (VEGF),

    that stimulate the growth o new blood vessels and also mark-edly increase retinal vascular permeability. The abnormal new

    vessels, associated fbrous tissue, and macular edema are major

    causes o the sight-threatening complications in diabetic eyedisease. By destroying a portion o the peripheral retina with

    laser, it has been hypothesized that retinal metabolic demandsand available nutrients are better balanced and the stimulus or

    growth o the new blood vessels is decreased. This treatment hasbeen termed panretinal photocoagulation (Fig. 39.15) and signif-

    cantly reduces the risk o vision loss due to neovascularization.

    Fig. 39.15 Fundus photograph o a patient 1 week ollowingapplication o panretinal photocoagulation with argon laser.

    retinal laser spot size the therapeutic window declines rom 5.4

    to 3.7 to 3.1 when pulse durations decrease rom 100 to 20 to10 ms, respectively. With both spot sizes, the therapeutic window

    decreases to unity as pulse durations decrease to 1 ms. At thispoint there is eectively no sae range o retinal photocoagula-

    tion: mild lesion and rupture are equally likely to occur at the

    same power.The width o the sae therapeutic window should sufce to

    accommodate or variations in undus pigmentation, which typi-

    cally do not exceed a actor o 2. To provide a sae therapeuticwindow larger than 2.5, pulse durations should equal or preer-ably exceed 10 ms or a beam o 330 m, and 20 ms or the

    132 m spot size.It is important to keep in mind that coagulation o blood

    vessels requires more energy than other tissue due to cooling bythe blood ow. For example, i a spot size o 200 m with expo-

    sure time o 200 ms is applied to occlude a blood vessel with

    ow velocity o 5 mm/s, the laser energy is eectively distrib-uted over the column fve times longer than the diameter o the

    laser spot. Thus the eective energy remaining at the photoco-agulation site is fve times lower than it would be in stationary

    tissue.

    Healing of retinal lesionsStudies in rabbits demonstrate that in photocoagulation lesionsthe RPE layer is restored within a week, though its pigmentation

    may remain abnormal either hyper- or hypopigmented.34 Inintense and moderate lesions gliotic scar flling the coagulated

    retinal layers stabilizes ater 1 month, and the wound contracts

    to approximately 40% o the original lesion diameter, as shownin the right column in Figure 39.11A and B. However, in very

    light lesions (barely visible clinical grade), photoreceptors con-tinue to shit into the damage zone and completely refll it by 4

    months, as shown in Figure 39.11C, in the right panel. As aresult, scarring and scotomata typically associated with conven-

    tional photocoagulation may be minimized or even completely

    avoided.

    34

    A similar phenomenon o restorative retinal plasticityhas recently been observed in rats35 and primates.36 It is impor-tant to keep in mind, though, that in order to maintain clinical

    efcacy o photocoagulation with smaller and lighter lesions, a

    larger number o them should be applied, to keep the same totalcoagulated area.33

    Pattern-scanning laser photocoagulationThe frst attempts to make photocoagulation a completely auto-

    mated procedure involved rather complex equipment, includingimage recognition sotware and eye tracking.37 The complexity

    o such systems prevented their commercial introduction andacceptance in clinical practice.

    A semiautomatic pattern-scanning photocoagulator (PASCAL,

    Topcon Medical Laser Systems) was introduced by OptiMedicain 2005.38 It delivered patterns o laser spots, ranging rom a

    single spot to 56 spots applied in a rapid sequence with a singledepression o a oot pedal. The control o laser parameters was

    perormed by means o a touch screen graphic user interace,acilitating selection o the dierent patterns o photocoagula-

    tion. The laser was activated by pressing a oot pedal, which waskept depressed until the entire pattern was completed, although

    it is possible or the physician to release the oot pedal and stopthe laser at will, prior to completion o the pattern, i clinically

    indicated.

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    11/15

    756

    Section

    4

    TranslationalBasicScience

    BasicS

    cience

    and

    Translation

    to

    Therapy

    Absorption o these wavelengths in macular pigments m

    cause heating and destruction o the nerve fber layer, resultiin loss o vision. As shown inFigure 39.1B, in the macular regi

    wavelengths longer than 500 nm should be chosen, such as tgreen argon (514 nm) or the requency-doubled YAG (532 n

    or semiconductor yellow (577 nm) laser. Melanin provides go

    absorption at most photocoagulation wavelengths. Wavelengselection is thereore less important when melanin is the prima

    absorber. To minimize scattering loss in cataract or in vitreo

    opacities the longer wavelengths (yellow, 577 nm, or r640680 nm) are efcacious. I scattering by the ocular tissuesnot signifcant, the argon green or doubled YAG continues

    serve well.When hemoglobin is the primary absorber (Fig. 39.1B), as

    the treatment o vascularized tumors, a wavelength shorter th600 nm is preerable. Treatment o CNV may be eective usi

    red light through indirect heat transer rom the surroundi

    melanin. In general, when photocoagulating structures conta large quantity o hemoglobin, wavelengths between 520 a

    580 nm are best suited. Ideally, or coagulation o blood vessthe photon penetration depth should be similar to the ves

    diameter, thus providing uniorm heating o the blood veswithout superfcial damage and peroration.

    Tunable lasers may provide the exibility to select a walength o choice or required photothermal procedure. Howev

    tunable lasers are more costly, require more maintenance, aare now less commonly employed clinically than previously.

    PhotodisruptionWhen tissue temperature exceeds the vaporization threshovapor bubbles are produced, which may lead to rupture o t

    tissue within a zone comparable to the bubble size. This proco explosive vaporization is typically employed or tissue diss

    tion. The actual temperature required or vaporization varbetween 100 and 305C depending on pulse duration and

    presence o the bubble nucleation sites.44 For efcient heatingtissue the pulse energy should be delivered ast enough to avo

    heat diusion rom the laser absorption zone during the pul

    a condition called thermal confnement. In other words, laser pulse duration should be shorter than the heat relaxati

    time, or the time o heat diusion rom the zone o laser absotion, L:

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    12/15

    to the tissue surace may produce a water jet which is capable o

    damaging tissue at distances exceeding the bubble radius by aactor o 4.46,47

    Explosive vaporization can be produced by absorption o laserradiation in water or in tissue. Strong absorption in water can

    be achieved at mid-inrared wavelengths. For example, penetra-

    tion o the Er:YAG laser (= 2.94 mm) in water is about 1 m.Shallow penetration o these lasers necessitates the fber-based

    delivery o this light into a liquid medium. A thin layer o water

    in ront o the intraocular probe is overheated with the laserpulse, and the resulting vaporization leads to rupture o tissuein proximity to the probe. The best implementation o this prin-

    ciple has been achieved with Er:YAG-based dissection o epireti-nal membranes48,49 (Fig. 39.18). Since a burst o closely spaced

    pulses, rather than a single pulse, was applied in that device, theactual vapor bubble had an elliptical shape and extended several

    hundreds o microns rom the probe.50

    Alternatively, overheating o liquid can be achieved with alaser strongly absorbed in the tissue constituents. For example,

    a fber-delivered ArF excimer laser ( = 193 nm), which isstrongly absorbed by proteins (penetration depth 0.2 m in

    generated which propagate with supersonic velocities and may

    result in signifcant damage to tissue, such as disruption andragmentation.44

    Vaporization o water in an overheated volume results in theormation o a short-living vapor bubble (a so-called cavitation

    bubble) which expands, cools down, and collapses during the

    time determined by its radius at maximal expansion. The lietimeo the spherical cavity with radius R0 in ree nonviscous liquid

    is described by the Rayleigh equation:

    = 0 91 0

    0

    . RP

    , where

    is liquid density and P0 is ambient pressure.45 For example, in

    water at atmospheric pressure, a cavity o 0.1 mm in radius col-

    lapses in about 10 s. (Growth and collapse o a spherical bubbletake approximately the same time.) Symmetric collapse o a

    spherical cavity may lead to overheating o the liquid and orma-tion o the secondary bubble. Due to the short lietime these

    bubbles are not visible to the eye during surgery, but they can

    be easily visualized using ast ash photography. As shown inFigure 39.17, at the liquidtissue interace the bubble might be

    deormed, and the secondary bubble may not be created. Impor-tantly, a collapsing cavitation bubble at the fber probe or next

    Fig. 39.17 Dynamics o the cavitation bubble at the gelatinsaline interace observed with ast ash photography. Bubble is created by a pulse oArF excimer laser delivered via the tapered optical fber. Numbers in each rame indicate a delay in microseconds between the laser pulse(10 ns) and a microsecond-long ash.

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    13/15

    758

    Section

    4

    TranslationalBasicScience

    BasicS

    cience

    and

    Translation

    to

    Therapy

    clinical studies have shown the efcacy o SRT in diabetic mac

    lopathy, central serous choroidopathy,62,63 and suboveal uater rhegmatogenous retinal detachment.64,65 Despite its clini

    promise, this technique has not been commercialized. One o tdifculties with SRT is the lack o visible change in the retin

    appearance, making it difcult to assess adequate laser do

    metry as the applications are being placed. An acousto-optisystem is currently under development that may help to ass

    the threshold energy density required or cavitation in RPE.66

    alternative approach to SRT is the rapid scanning o a laser be

    providing microsecond exposures, sufciently short or selecttreatment o the RPE.67,68

    FUTURE DEVELOPMENTS

    Monitoring retinal temperatureRetinal thermal therapies with temperature rise below t

    threshold o immediately visible tissue response, such as sulethal hyperthermia, do not have as high a degree o predi

    ability as conventional thermal photocoagulation methods. Fsuccessul sublethal treatment the temperature rise in tissue

    on the order o 10C.69 However, due to the strong variationundus pigmentation the light absorption varies rom patient

    patient, and even between dierent areas in the same eye. The

    ore the same irradiation settings may lead to very diereresults in dierent patients, and so direct measurement o reti

    temperature during such treatments is highly desirable. Simlarly, it would be desirable to monitor retinal temperature in

    treated spot during photocoagulation to provide uniorm ocomes in areas with dierent pigmentation.

    A noninvasive method o determination o retinal temperatuhas been developed, which is based on the detection o acous

    waves generated in RPE by short laser pulses.70 An acoustransducer or the detection o the pressure waves is built int

    contact lens attached to the treated eye during the procedu

    The pressure waves are generated due to thermoelastic expsion o melanosomes upon absorption o the short (submicros

    ond) laser pulses. The key issue in this approach is that thermoelastic expansion coefcient o water var

    tissue), has been applied or dissection o epiretinal and subreti-nal membranes.51,52 In this case the laser light overheats the tissue

    and leads to vaporization o its water content with subsequent

    tissue rupturing.53,54 Despite the early promise o both o thesedevices (Er:YAG and ArF excimer lasers) in clinical tests, both

    have ailed to achieve widespread acceptance in medical practicedue in part to cost, the rigidity o the fbers, and the lack o

    coagulation capability.Another approach to dissection o transparent tissue utilizes

    ionization o the material and ormation o plasma rom a high-intensity laser beam. At extremely high irradiances (108-1011 W/

    cm2), that can be achieved in a short-pulsed (nss) tightlyocused laser beam, transparent material can be ionized and ions

    absorbing the laser light reach very high temperatures.55 This

    mechanism, called dielectric breakdown, allows or a very local-ized deposition o energy in the middle o a transparent liquid

    or solid at the ocal point o the laser beam. This process iswidely used in ragmentation o the opacifed posterior lens

    capsule (secondary cataract) with nanosecond Nd:YAG lasers.At shorter pulse durations (1 ps100 s) and lower energies, this

    process is applied to intrastromal ablation ormation o acorneal ap or reractive surgery.56,57 This approach has also

    been tested in the dissection o epiretinal membranes using the

    tightly ocused beam directed rom outside the eye.58 Despite theact that very low energy is required or this process (several

    microjoules with ps lasers), its applicability in the posterior poleis limited due to the difculty in axial dierentiation between

    the epiretinal membranes and the retina located very closebehind them. In addition, strong optical aberrations in the

    periphery o the posterior pole preclude tight ocusing o thelaser beam in these areas.

    Selective retina therapy (SRT)Light is strongly absorbed in the melanosomes in the RPE (a

    8000 cm-1).59 The application o short (submicrosecond) laserpulses allows or confnement o the thermal and mechanical

    eects o this absorption within the RPE layer, thus sparing the

    photoreceptors and the inner retina60,61 (Fig. 39.19). It has beendemonstrated that the application o repetitive pulses o micro-

    second and submicrosecond duration results in selective damageto RPE, presumably due to the ormation o small cavitation

    bubbles around melanosomes.59 Subsequent RPE prolierationand migration restore continuity o the RPE layer. Several small

    Fig. 39.18 Epiretinal membrane dissected by the Er:YAG laser. Notethe absence o retinal damage despite its close proximity.

    Fig. 39.19 Rabbit retina 24 hours ater selective retina therapy withinrared semiconductor laser. Note that damage is almost exclusivelyconfned to the retinal pigment epithelial (RPE) layer with preservatioo the outer nuclear layer and even the outer segments o overlyingphotoreceptors. There is a small localized eusion between the RPEand photoreceptors.

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    14/15

    neovascularization-verteporfn in photodynamic therapy report 2. Am JOphthalmol 2001;131:54160.

    17. Luttrull JK, Musch DC, Mainster MA. Subthreshold diode micropulse photo-coagulation or the treatment o clinically signifcant diabetic macular oedema.Br J Ophthalmol 2005;89:7480.

    18. Figueira J, Khan J, Nunes S, et al. Prospective randomised controlled trialcomparing sub-threshold micropulse diode laser photocoagulation andconventional green laser or clinically signifcant diabetic macular oedema.Br J Ophthalmol 2009;93:13414.

    19. Luttrull JK, Musch DC, Mainster MA. Subthreshold diode micropulse photo-coagulation or the treatment o clinically signifcant diabetic macular oedema.Br J Ophthalmol 2005;89:7480.

    20. Luttrull JK, Musch DC, Spink CA. Subthreshold diode micropulse panretinalphotocoagulation or prolierative diabetic retinopathy. Eye (Lond) 2008;22:60712.

    21. Ohkoshi K, Yamaguchi T. Subthreshold micropulse diode laser photocoagula-tion or diabetic macular edema in Japanese patients. Am J Ophthalmol 2010;149:1339.

    22. Desmettre TJ, Mordon SR, Buzawa DM, et al. Micropulse and continuouswave diode retinal photocoagulation: visible and subvisible lesion parameters.Br J Ophthalmol 2006;90:70912.

    23. Luttrull JK, Sramek C, Palanker D, et al. Long-term saety, high-resolutionimaging, and tissue temperature modeling o sub-visible diode micropulsephotocoagulation or retinovascular macular edema. Retina 2011;epub aheado print.

    24. Niemz M. Lasertissue interactions. Fundamentals and applications. Berlin:Springer; 2002.

    25. Sramek C, Paulus Y, Nomoto H, et al. Dynamics o retinal photocoagulationand rupture. J Biomed Optics 2009;14:03400713.

    26. Oosterhuis JA, Journeedekorver HG, Kakebeekekemme HM. Transpupillarythermotherapy in choroidal melanomas. Arch Ophthalmol 1995;113:693693.

    27. Reichel E, Berrocal AM, Ip M, et al. Transpupillary thermotherapy o occultsuboveal choroidal neovascularization in patients with age-related macular

    degeneration. Ophthalmology 1999;106:190814.28. Newsom RSB, McAlister JC, Saeed M, et al. Transpupillary thermotherapy

    (TTT) or the treatment o choroidal neovascularisation. Br J Ophthalmol2001;85:1738.

    29. Svaasand LO. Laser-induced hyperthermia physics considerations andlimitations. Laser Surg Med 1988;8:182182.

    30. Svaasand LO, Gomer CJ, Profo AE. Laser-induced hyperthermia o oculartumors. Appl Optics 1989;28:22807.

    31. Mainster MA, Reichel E. Transpupillary thermotherapy or age-related maculardegeneration: Long-pulse photocoagulation, apoptosis, and heat shock pro-teins. Ophthalmic Surg Las 2000;31:35973.

    32. Jain A, Blumenkranz MS, Paulus Y, et al. Eect o pulse duration on size andcharacter o the lesion in retinal photocoagulation. Arch Ophthalmol2008;126:7885.

    33. Palanker D, Lavinsky D, Blumenkranz MS, et al. The impact o pulse durationand burn grade on size o retinal photocoagulation lesion: implications orpattern density. Retina 2011;31:16649.

    34. Paulus YM, Jain A, Gariano RF, et al. Healing o retinal photocoagulationlesions. Invest Ophthalmol Vis Sci 2008;49:55405.

    35. Belokopytov M, Belkin M, Dubinsky G, et al. Development and recovery olaser-induced retinal lesion in rats. Retina 2010;30:66270.36. Merigan WH, Strazzeri J, DiLoreto DA, Jr, et al. Visual recovery ater outer

    retinal damage in the macaque. Invest Ophthalmol Vis Sci 2011;52:3202.37. Wright CHG, Barrett SF, Ferguson RD, et al. Initial in vivo results o a hybrid

    retinal photocoagulation system. J Biomed Opt 2000;5:5661.38. Blumenkranz MS, Yellachich D, Andersen DE, et al. Semiautomated patterned

    scanning laser or retinal photocoagulation. Retina 2006;26:3706.39. Nagpal M, Marlecha S, Nagpal K. Comparison o laser photocoagulation or

    diabetic retinopathy using 532-nm standard laser versus multispot pattern scanlaser. Retina 2010;30:4528.

    40. Muqit MM, Marcellino GR, Gray JC, et al. Pain responses o Pascal 20 ms multi-spot and 100 ms single-spot panretinal photocoagulation: Manchester PascalStudy, MAPASS report 2. Br J Ophthalmol 2010;94:14938.

    41. Muqit MM, Gray JC, Marcellino GR, et al. In vivo lasertissue interactions andhealing responses rom 20- vs 100-millisecond pulse Pascal photocoagulationburns. Arch Ophthalmol 2010;128:44855.

    42. DRS Study Group. Photocoagulation treatment or prolierative diabeticretinopathy. clinical application o DRS fndings, DRS report number 8.Ophthalmology 1981;88:583600.

    43. ETDRS Study Group. Early Photocoagulation or Diabetic Retinopathy. ETDRSreport number 9. Ophthalmology 1991;98:76685.

    44. Vogel A, Venugopalan V. Mechanisms o pulsed laser ablation o biologicaltissues. Chem Rev 2003;103:20792079.

    45. Young FR. Cavitation. Maidenhead: McGraw-Hill; 1989. p. 136.46. Palanker D, Vankov A, Miller J. Eect o the probe geometry on dynamics o

    cavitation. SPIE, LaserTissue Interaction XIII 2002;4617:1127.47. Palanker D, Vankov A, Miller J, et al. Prevention o tissue damage by water jet

    during cavitation. J Appl Phys 2003;94:265461.48. DAmico DJ, Blumenkranz MS, Lavin MJ, et al. Multicenter clinical experience

    using an erbium:YAG laser or vitreoretinal surgery. Ophthalmology1996;103:157585.

    49. DAmico DJ, Brazitikos PD, Marcellino GR, et al. Initial clinical experience withan erbium:YAG laser or vitreoretinal surgery. Am J Ophthalmol 1996;121:41425.

    with temperature, by about 1% per 1C.70 This eect allows or

    monitoring the changes in temperature o the RPE cells by moni-toring the changes in amplitude o acoustic waves generated by

    the laser pulses o constant energy. The probing laser pulses areapplied simultaneously with application o a therapeutic laser

    to detect temperature rise in tissue during the exposure. It has

    been demonstrated that precision o this method is on the ordero 1C. Clinical testing o the system is currently in progress.

    Optical monitoring of tissue changes

    in real timeAn optical approach to real-time eedback during retinal photo-

    coagulation has recently been demonstrated.71 It is based on OCTmonitoring o the heated tissue expansion and changes in retinal

    scattering during coagulation. The system operates with ms tem-poral resolution, which should be ast enough or real-time

    monitoring o retinal photocoagulation.Another technique or detection o tissue condition during

    slow thermal therapy is based on spectroscopy o white lightscattered rom the tissue.72 Cellular response to thermal stress

    involves expression o various proteins, as well as changes in

    their aggregation and concentration. All these eects result inchanges o the reractive indices and/or the sizes and shapes o

    the cellular organelles, which can be detected using light-scattering spectroscopy. Particle sizes down to 100 nm in diam-

    eter can be detected using light within the spectral range o3501000 nm.72 Since the inormation is obtained optically and

    without any staining this technique operates in real time and isnoninvasive. It has been observed that scattering coefcients o

    some organelles change very strongly (up to 70%) and rapidly(20 seconds) in the heated cells.73

    REFERENCES1. Meyer-Schwickerath G. Light coagulation. St Louis: Mosby; 1960.2. Palanker DV, Blumenkranz MS, Marmor MF. 50 years o ophthalmic laser

    therapy. Arch Ophthalmol 2011;129:16139.3. Smith G, Atchinson DA. The eye and visual optical instruments. Cambridge:

    Cambridge University Press; 1996, chapter 13.

    4. Thompson KP, Ren QS, Parel J-M. Therapeutic and diagnostic application olasers in ophthalmology. In: Waynant RW, editor. Lasers in medicine.Boca Raton FL: CRC Press; 2002, chapter 8.

    5. Pomerantze O, Pankratov M, Wang GJ, et al. Wide-angle optical-model o theeye. Am J Optom Phys Opt 1984;61:16676.

    6. Walsh G, Charman WN, Howland HC. Objective technique or the determina-tion o monochromatic aberrations o the human eye. J Opt Soc Am A 1984;1:98792.

    7. Doornbos RMP, Lang R, Aalders MC, et al. The determination o in vivo humantissue optical properties and absolute chromophore concentrations using spa-tially resolved steady-state diuse reectance spectroscopy. Phys Med Biol1999;44:96781.

    8. Troy TL, Thennadil SN. Optical properties o human skin in the near inraredwavelength range o 1000 to 2200 nm. J Biomed Opt 2001;6:16776.

    9. Dougherty TJ, Mang TS. Characterization o intra-tumoral porphyrin ollowinginjection o hematoporphyrin derivative or its purifed component. PhotochemPhotobiol 1987;46:6770.

    10. Schmidt U, Birngruber R, Hasan T. [Selective occlusion o ocular neovascular-ization by photodynamic therapy.] Ophthalmologe 1992;89:3914.

    11. Miller H, Miller B. Photodynamic therapy o subretinal neovascularization in

    the monkey eye. Arch Ophthalmol 1993;111:85560.12. Kramer M, Miller JW, Michaud N, et al. Liposomal benzoporphyrin derivative

    verteporfn photodynamic therapy. Selective treatment o choroidal neovascu-larization in monkeys. Ophthalmology 1996;103:42738.

    13. Miller JW, Walsh AW, Kramer M, et al. Photodynamic therapy o experimentalchoroidal neovascularization using lipoprotein-delivered benzoporphyrin.Arch Ophthalmol 1995;113:8108.

    14. Woodburn KW, Engelman CJ, Blumenkranz MS. Photodynamic therapy orchoroidal neovascularization: a review. Retina 2002;22:391405;.

    15. Birngruber R, Indor L, Soultanopoulos D, et al. Photodynamic occlusion oocular neovascularization: Preclinical evaluation o liposomal zinc phthalocya-nine. Invest Ophthalm Vis Sci 1996;37:42144214.

    16. Arnold J, Kilmartin D, Olson J, et al. Verteporfn therapy o suboveal choroidalneovascularization in age-related macular degeneration: Two-year results o arandomized clinical trial including lesions with occult with no classic choroidal

  • 7/27/2019 Retinal Laser Therapy Biophysical Basis

    15/15

    760

    Section

    4

    TranslationalBasicScience

    BasicS

    cience

    and

    Translation

    to

    Therapy

    63. Elsner H, Porksen E, Klatt C, et al. Selective retina therapy in patients wcentral serous chorioretinopathy. Graees Arch Clin Exp Ophthal2006;244:163845.

    64. Koinzer S, Elsner H, Klatt C, et al. Selective retina therapy (SRT) o chrosuboveal uid ater surgery o rhegmatogenous retinal detachment: three creports. Graees Arch Clin Exp Ophthalmol 2008;246:13738.

    65. Brinkmann R, Roider J, Birngruber R. Selective retina therapy (SRT): a revon methods, techniques, preclinical and frst clinical results. Bull Soc BeOphtalmol 2006;5169.

    66. Brinkmann R, Schuele G, Joachimmeyer E, et al. Determination o absoundus temperatures during retinal laser photocoagulation and selective Rtreatment. Invest Ophthalmol Vis Sci 2001;42:S696S696.

    67. Framme C, Alt C, Schnell S, et al. Selective targeting o the retinal pigmepithelium in rabbit eyes with a scanning laser beam. Invest Ophthalmol Sci 2007;48:178292.

    68. Paulus YM, Jain A, Nomoto H, et al. Selective retinal therapy with microsecexposures using a continuous line scanning laser. Retina 2011. 31:3808.

    69. Sramek C, Mackanos M, Spitler R, et al. Non-damaging retinal phototherdynamic range o heat shock protein expression. Invest Ophthalmol Vis2010;52:17807.

    70. Schule G, Huttmann G, Framme C, et al. Noninvasive optoacoustic tempture determination at the undus o the eye during laser irradiation. J BiomOpt 2004;9:1739.

    71. Huttmann G, Mller H, Schlott K, et al. Investigating o retinal photocoagtion by high-speed OCT in rabbits. Invest Ophthalmol Vis Sci 2011549.

    72. Fang H, Ollero M, Vitkin E, et al. Noninvasive sizing o subcellular organewith light scattering spectroscopy. IEEE J Sel Top Quant 2003;9:26776.

    73. Schuele G, Huie P, Vankov A, et al. Non-invasive monitoring o the therstress in RPE using light scattering spectroscopy. Ophthalm Technol SProc 2004;5314.

    50. Lin CP, Stern D, Puliafto CA. High-speed photography o Er: YAG laser abla-tion in uid. Implication or laser vitreous surgery. Invest Ophthalmol Vis Sci1990;31:254650.

    51. Palanker D, Hemo I, Turovets I, et al. Vitreoretinal ablation with the 193-nmexcimer laser in uid media. Invest Ophthalmol Vis Sci 1994;35:383540.

    52. Hemo I, Palanker D, Turovets I, et al. Vitreoretinal surgery assisted by the193-nm excimer laser. Invest Ophthalmol Vis Sci 1997;38:18259.

    53. Palanker D, Turovets I, Lewis A. Mechanisms o tissue damage during ArFexcimer endolaser microsurgery. LaserTissue Interaction VII, Proc SPIE1996;2681:2205.

    54. Palanker D, Turovets I, Lewis A. Dynamics o ArF excimer laser-induced cavi-tation bubbles in gel surrounded by a liquid medium. Lasers Surg Med1997;21:294300.

    55. Vogel A, Busch S, Jungnickel K, et al. Mechanisms o intraocular photodisrup-tion with picosecond and nanosecond laser pulses. Lasers Surg Med1994;15:3243.

    56. Krueger RR, Quantock AJ, Juhasz T, et al. Ultrastructure o picosecond laserintrastromal photodisruption. J Reract Surg 1996;12:60712.

    57. Yen KG, Sachs Z, Elner VE, et al. Histopathology o emtosecond laser intra-stromal reractive surgery in rabbits. Invest Ophthalmol Vis Sci 1999;40:S621S621.

    58. Cohen BZ, Wald KJ, Toyama K. Neodymium:YLF picosecond laser segmenta-tion or retinal traction associated with prolierative diabetic retinopathy.Am J Ophthalmol 1997;123:51523.

    59. Brinkmann R, Huttmann G, Rogener J, et al. Origin o retinal pigment epithe-lium cell damage by pulsed laser irradiance in the nanosecond to microsecondtime regimen. Laser Surg Med 2000;27:45164.

    60. Roider J, Hillenkamp F, Flotte T, et al. Microphotocoagulation: selective eectso repetitive short laser pulses. Proc Natl Acad Sci U S A 1993;90:86437.

    61. Roider J, Michaud NA, Flotte TJ, et al. Response o the retinal pigment epithe-lium to selective photocoagulation. Arch Ophthalmol 1992;110:178692.

    62. Roider J, Brinkmann R, Wirbelauer C, et al. Subthreshold (retinalpigment epithelium) photocoagulation in macular diseases: a pilot study.

    Br J Ophthalmol 2000;84:407.