Ions for Non Malignancies

download Ions for Non Malignancies

of 12

Transcript of Ions for Non Malignancies

  • 7/29/2019 Ions for Non Malignancies

    1/12

    Particle therapy for noncancer diseases

    Christoph BertGSI Helmholtzzentrum fur Schwerionenforschung, Biophysics Department, Planckstrae 1, 64291 Darmstadt,Germany

    Rita Engenhart-CabillicPhilipps-University Marburg, Center for Radiology, Department of Radiation Therapy, Baldinger Strasse,35043 Marburg, Germany

    Marco Durantea)

    GSI Helmholtzzentrum fur Schwerionenforschung, Biophysics Department, Planckstrae 1, 64291 Darmstadt,Germany; Technische Universitat Darmstadt, Institut fur Festkorperphysik, Hochschulstrae 3, 64289Darmstadt, Germany; and Frankfurt Institute for Advanced Studies, Johann Wolfgang Goethe University,Ruth-Moufang-Str. 1, 60438 Frankfurt am Main, Germany

    (Received 3 August 2011; revised 3 January 2012; accepted for publication 13 February 2012;published 8 March 2012)

    Radiation therapy using high-energy charged particles is generally acknowledged as a powerful new

    technique in cancer treatment. However, particle therapy in oncology is still controversial, specifically

    because it is unclear whether the putative clinical advantages justify the high additional costs.

    However, particle therapy can find important applications in the management of noncancer diseases,

    especially in radiosurgery. Extension to other diseases and targets (both cranial and extracranial) may

    widen the applications of the technique and decrease the cost/benefit ratio of the accelerator facilities.Future challenges in this field include the use of different particles and energies, motion management

    in particle body radiotherapy and extension to new targets currently treated by catheter ablation (atrial

    fibrillation and renal denervation) or stereotactic radiation therapy (trigeminal neuralgia, epilepsy,

    and macular degeneration). Particle body radiosurgery could be a future key application of

    accelerator-based particle therapy facilities in 10 years from today. VC 2012 American Association of

    Physicists in Medicine. [http://dx.doi.org/10.1118/1.3691903]

    I. BACKGROUND AND INTRODUCTION

    With the increasing number of accelerator facilities for parti-

    cle therapy currently planned, proposed, or under construc-

    tion, the debate on the cost/benefit ratio is now going beyondthe radiotherapy community and involving the general pub-

    lic, with several reports in the press. Therapy with acceler-

    ated charged particles exploits protons or heavier ions

    (typically carbon) produced by accelerators.15 Current clini-

    cal results support the predictions that charged particles are

    effective in tumor killing with greater sparing of the normaltissue, including a reduced risk of secondary cancers, which

    makes it particularly important for pediatric cancers.6,7 How-

    ever, direct clinical trials comparing x- or c-rays to protons

    or carbon ions are still lacking,8 and this fuels the contro-

    versy on the cost/benefit ratio of ion therapy.

    Since the first patient treatments in the Lawrence Berke-

    ley Laboratory (USA) and Uppsala (Sweden) at the end of

    the 1950s,9 almost 100 000 patients have been treated with

    energetic ions all over the world for different cancers. Over

    80% of these patients were treated with protons.10 The cost

    of particle therapy is dominated by the construction cost,

    which currently ranges from $35 M$ (for a single room,proton therapy commercial machine) up to $350 M$ (for a

    heavy ion center including radiodiagnostic and conventional

    x-ray therapy accelerators such as the one in construction in

    Shanghai, China). In the United States, where heavy ion

    therapy started in Berkeley, particle therapy is limited to pro-

    tons, with eight centers currently in operation. Many new

    centers have been proposed, but at the moment, only a hand-

    ful of facilities have the capability of treating patients with

    different ions (see Sec. II A and Ref. 10). The main problem

    is that the cost of particle therapy is more than two times

    higher than the most advanced x-ray methods.8 Of course,cost-effectiveness is more than just looking at the costs: so-

    phisticated model-based economic evaluations, including in

    silico clinical trials, are necessary to provide sensible advi-

    ces for medical decision-making.11 Independent evidence-

    based studies on the cost-effectiveness of the particle therapy

    have indeed concluded that the current lack of evidence forbenefit of protons should provide a stimulus for continued

    research.12 In this paper, we provide some new research

    topics that can potentially boost the benefits of the particle

    therapy centers.

    One factor that is generally not considered in planning

    particle facilities is the possibility of applications in non-

    cancer diseases. The charged particle depth-dose distribution

    is completely different from x-rays, c-rays, electrons, or neu-

    trons. Most of the energy is released in a narrow volume at

    the end of the particle path in the body, the Bragg peak (Fig.

    1). The relative straggling is proportional to $M1/2, and the

    lateral beam spread to $1/bpc, where Mis the particle mass,p the momentum and b v/c the relative velocity.5 As a con-

    sequence, the Bragg peak can become more and more nar-

    row using heavy ions (i.e., increasing Mcompare protons

    to C-ion Bragg peaks at the same range in Fig. 1) and

    increasing the particle energy (i.e., increasing b). Eventually,

    the Bragg peak can be used as a remote scalpel for

    1716 Med. Phys. 39 (4), April 2012 0094-2405/2012/39(4)/1716/12/$30.00 VC 2012 Am. Assoc. Phys. Med. 1716

  • 7/29/2019 Ions for Non Malignancies

    2/12

    radiosurgery. Stereotactic radiosurgery (SRS) or stereotactic

    radiation therapy (SRT) (Ref. 13) are successfully performed

    in many clinical centers using x- or c-rays (e.g., Cyberknife

    and Gammaknife) for several noncancer disorders such as

    trigeminal neuralgia, arterial aneurism, and arteriovenous

    malformations (AVM). In principle, the narrow Bragg peakcould be used more successfully for these and other non-

    cancer disorders. The number of noncancer diseases eligible

    for particle radiosurgery can be drastically increased if mov-

    ing organs are considered. Management of organ motion is

    indeed a problem both for x-ray14 and particle15 radiother-apy, but recent progress in the field suggest that, in the com-

    ing 10 years, the problem may be tackled with sufficient

    precision (see Sec. II D).

    II. CURRENT STATE OF THE ART

    II.A. Ion therapy in oncology

    The current status of particle therapy in oncology has

    been reviewed several times in the past few years, including

    the Vision 20/20 series on this Journal.2,3 For this review, it

    is enough to remark that proton therapy is now a widely

    adopted conformal radiotherapy method, and despite the

    controversy on the cost/benefit ratio compared to conven-

    tional x-ray therapy,12 it is currently used even in the ab-

    sence of phase III clinical trials.16 For heavy-ion therapy, the

    experience is more limited, but the advantages compared to

    x-rays are potentially much greater, thanks to the differentbiological effects in the Bragg peak region.1 Clinical trials

    comparing heavy ions to protons or photons are complicated

    by the relative paucity of patients and differences in fractio-

    nation scheme and LET.17 In addition to the large set of clin-

    ical data already acquired at NIRS (Japan) with C-ions,4

    more clinical results on different cancers treated with pro-

    tons and carbon ions are coming from the new Heidelberg

    facility (HIT) (Refs. 16 and 17) in Germany. The new center

    in Italy (CNAO) started treatments in 2011 and will treat

    patients with both H- and C-ions. Several new facilities with

    capabilities to deliver both H- and C-ions are currently under

    construction in Europe and Asia.10 Access to proton or heav-

    ier ions facilities is therefore becoming easier and should

    allow tests in noncancer diseases.

    II.B. Stereotactic particle radiotherapy andradiosurgery

    Considering the favorable dose-depth distribution com-pared to photons, particle therapy in oncology is often eval-

    uated in competition to stereotactic x-ray therapy. The word

    stereotactic refers to the use of a 3D coordinates system to

    locate small targets in the patient. Radiosurgery was indeed

    initially performed with protons18 and then with the Gamma-

    knife19 at the Karolinska Institute in Sweden. Latest versions

    include the use of special LINACs for pencil beam radiosur-

    gery (Cyberknife) and intensity modulated radiation therapy

    delivery (Tomotherapy).20 SRS is generally delivered in sin-

    gle fractions, while SRT is used for fractionated intracranialtreatments. Energetic charged particles have been used since

    many years in the treatment of AVM, but so far, very fewother skull or body lesions have been treated with protons or

    heavier ions.

    II.B.1. Treatment of AVM

    The potential of stereotactic particle radiosurgery (SpRS)

    or stereotactic particle radiotherapy (SpRT) was soon recog-nized in neurosurgery. For deep, inoperable AVM, SpRS/T

    represents an interesting alternative: when hemodynamic

    flow in AVM is different from normal vessels, focal beam

    irradiation of the shunting vessels leads to thrombosis andhemostasis, with eventual complete obliteration of the

    AVM. Treatment of AVM with protons has been one of the

    first noncancer applications of charged particle therapy21 and

    is indeed currently performed in several proton therapy cen-

    ters.22,23 At MGH-Francis H. Burr Proton Therapy Center

    (USA), the patient is placed in an immobilizing head frame

    on a couch that can be rotated around a fixed proton beam

    line. Using 3 linear and 2 rotational degrees of freedom, this

    system (known as the STAR device) is able to accurately

    position the brain lesions at the beam isocenter.24 An isocen-

    tric gantry can be used if more angles are needed in the treat-

    ment plan. Apart from protons, helium ions were used in the

    1950s at the Lawrence Berkeley National Laboratory.25

    A detailed comparison of protons vs x-rays for AVM

    treatment is complicated by the fact that, in most cases, pro-

    tons are used for large and irregularly shaped lesions,24

    which are very difficult to treat with photons but whoseprognosis is worse than for small AVM. Broadly speaking,

    protons seem to be comparable to photons and a better alter-

    native for large, irregular lesions. The experience at the pro-

    ton center in Uppsala (Sweden) shows that protons provide

    an advantage for lesions >10 ml.26 Results from Berkeley

    with He-ions demonstrated complete obliteration for small

    (25 ml)

    FIG. 1. Depth-dose distribution for high-energy x-rays, protons, and C-ionsat two different energies in tissue. For charged particles, most of the dose isdeposited in the distal region (the Bragg peak), and the beam penetrationdepth can be modified by changing the beam energy (particle velocity). TheBragg peak is broadened both in depth and in the plane for protons com-

    pared to carbons, because of their lower mass (courtesy of Dr. Uli Weber,Phillips University Marburg, Germany).

    1717 Bert, Engenhart-Cabillic, and Durante: Particle therapy for noncancer d iseases 1717

    Medical Physics, Vol. 39, No. 4, April 2012

  • 7/29/2019 Ions for Non Malignancies

    3/12

    lesions.27 A recent analysis of 64 patients treated with hypo-

    fractionated (23 fractions) protons at iThemba in South

    Africa28 shows obliteration in 67% of the patients with

    lesions smaller than 14 ml and 43% obliteration in patients

    with larger AVM volumes. Grade IV acute complications

    were observed in 3% of the patients.

    II.B.2. Stereotactic radiotherapy in other noncancerdiseases

    X- orc-ray SRS or SRT is currently under study for treat-

    ing several noncancer diseases in the skull including trigemi-

    nal neuralgia,29 epilepsy,30 intracranial aneurysm,31 and

    macular degeneration.32 These treatments may greatly

    expand the spectrum of skull lesions where external beam

    radiation therapy can replace surgery. In fact, trigeminal

    neuralgia (Fothergills disease ortic douloureux) has an inci-

    dence of 1 in 15 000 people, while macular degenerationaffects from 10% to 30% of the population older than 65

    years, representing the major cause of visual impairment in

    elderly. Results of SRS or SRT treatments using Gamma-knife or Cyberknife are generally considered positive,

    although randomized clinical trials are generally lacking.33

    Trigeminal neuralgia, which is characterized by a tempo-

    rary paroxysmal lancinating facial pain, can be treated with

    drugs (anticonvulsant or antidepressant) or alternatively with

    microvascular decompression, a craniotomy where the sur-

    geon has to locate and separate veins and arteries in contact

    with the trigeminal nerve.34 In fact, the pain is often caused

    by a blood vessel compressing or pulsating on the trigeminalnerve. In addition to external beam therapy, a further option

    is radiofrequency gangliotomy. SRS has been proven to be

    safe and effective in patients with medical- and surgical-

    refractory trigeminal neuralgia. In many cases, SRS is per-formed with a shot size around 5 mm, a maximum point

    dose of 8090 Gy and about 40 Gy prescribed at the 50%

    isodose line.35 If the patient is nonresponsive or the pain

    returns, SRS can be repeated, and up to 4 repeated Gamma-

    knife treatments have been reported in a few patients. The

    main treatment-related complication is facial numbness. The

    complication rate depends on the total dose and could be

    increased by repeated treatments.36

    For epilepsy patients nonresponsive to anticonvulsants,

    SRS is an alternative to resective surgery.37 Hypothalamic

    hamartomas and mesial temporal lobe epilepsy are small

    enough (50% isodose volume 50% of the patients lost >2 lines of vision. Improvements

    can be expected if different ions and energies can provide

    additional benefit, in terms of increased relative biological

    effectiveness (RBE) (see Sec. III A 2) or increased precision

    (see Sec. III A 1). Moreover, in the treatment of macular

    degeneration, ionizing radiation will be always associated to

    anti-VEGF drugs, and synergistic effects are possible.43 In

    vitro studies are necessary to compare protons and x-rayscombined to antiangiogenic drugs in cultures representative

    of the target tissue. Cultures of choroid endothelial cells mayrepresent an interesting in vitro model, and survival and pro-

    liferation of these cells after exposure to protons has been

    recently measured.44

    II.C. Stereotactic body radiation therapy

    Stereotactic body radiation therapy (SBRT) and stereotac-

    tic body particle radiation therapy (SBpRT) are generally

    used for treating small extracranial targets primarily in thorax

    [e.g., nonsmall cell lung cancers (NSCLC)]33,45,46 and abdo-

    men (e.g., hepatocellular carcinoma47,48), using hypofractio-

    nation. Different instrumentations are currently used in

    clinical practice including Cyberknife, Clinac (Novalis

    Shaped Beam), Tomotherapy, the Synergy System (Elekta,

    Stockholm, Sweden), and scattered particle beams. From 1 to

    12 radiation beams are used, generally in 15 fractions. Dif-

    ferent body immobilization systems are necessary, includingthe Alpha Cradle and the Stereotactic Body Frame.

    No direct comparative studies have been reported so far

    comparing SBRT to conformal fractionated radiotherapy,

    because they are used for different conditions: SBRT

    requires small well defined targets with large dose fractiona-

    tion, and conformal fractionated therapy is typically used for

    larger targets with standard fractionation. For stage I

    1718 Bert, Engenhart-Cabillic, and Durante: Particle therapy for noncancer d iseases 1718

    Medical Physics, Vol. 39, No. 4, April 2012

  • 7/29/2019 Ions for Non Malignancies

    4/12

    peripheral NSCLC treatment using C-ions, NIRS in Japan is

    performing an interesting phase I/II dose escalation study for

    several fractionation schemes: from 16 fractions in 6 weeks

    already in 1994, down to 4 fractions in 1 week since 2001,

    and finally a single fraction since 2004, i.e., stereotactic

    body particle radiosurgery (SBpRS).49

    Apart from tumor treatments, an interesting noncancer

    application of SBRT has been recently proposed: treatment

    of atrial fibrillation. This treatment has not yet reached the

    clinic and will be discussed in Sec. III B 1.

    II.D. Management of organ motion in SBRT

    Radiosurgery of extracranial lesions can only be per-

    formed taking into account the organ motion, as organ

    movement is already a severe hindrance in the fractionated

    high-dose regime.14 In particle beam therapy with a scattered

    beam, most centers use beam gating along with dedicateddefinition of the planning target volume to incorporate range

    changes caused by organ motion. For scanned particle ther-

    apy,15

    where the lesion is irradiated in the narrow particleBragg peak of small diameter, the problem is even more

    acute due to interference effects of scanned beam and mov-

    ing target.

    From a geometrical point of view, the impact of organ

    motion in particle therapy is comparable to the impact in 3D

    conformal photon beam therapy, i.e., target motion results in

    blurring of the dose gradients from target volume to normal

    tissue.14 In addition to these geometrical effects, the motion

    of organs in the target area and within the beams path canchange the radiological pathlength and thus the distribution

    of the deposited dose.50 These changes are independent of

    the delivery technique. For beam scanning, there are two

    additional interference options: one is due to interfieldmotion in intensity modulated particle therapy (IMPT), the

    other due to interference between scanning motion and intra-

    fractional (or intrafield) organ motion, which is often called

    interplay and typically results into underdosage and overdos-

    age within the target volume (Fig. 2).15 As we will discuss

    below, options for SBpRS heavily rely on management of

    organ motion, in particular if used in combination with a

    scanned beam that provides best conformation. Especially

    for treatment of cardiac lesions, new techniques will be

    required since the heart is not only beating but also moving

    due to respiration.

    Current management of inter- and intrafractional motion in

    charged particle beam therapy is widely based on the methods

    established for x-ray therapy.15 A distinction should be made

    with respect to the beam delivery method, i.e., whether pas-

    sive scattering (e.g., MGH or Loma Linda in USA) or active

    scanning (e.g. PSI, Switzerland or HIT, Germany) are used. Inscattered beam treatments, the use of margins is sufficient to

    ensure dose coverage of the clinical target volume (CTV) if

    the range influence is addressed. For prostate treatments (as

    an example for interfractional motion51) as well as lung can-

    cer treatments (as an example for intrafractionally moving

    organs52), the use of margins is the dominant technique, sup-

    plemented by gating in case of large respiratory motion ampli-

    tudes.53 Gating is the method used at NIRS in Japan for the

    SBpRS of NSCLC (see Sec. II C). For prostate treatments, the

    potential intrafractional motion components are often sup-

    pressed by dedicated immobilization, i.e., controlled rectum

    and bladder filling by drinking schemes, enemas, or rectal bal-

    loons.54 Range uncertainties can be addressed by compensator

    smearing and/or manual changes of the underlying planning

    CT scan (which typically is a free-breathing or a gated CT ex-

    amination55,56). These changes aim of replacing low-density

    lung tissue by high-density tumor tissue to ensure proper tar-

    get coverage in all states of the respiratory cycle. For patient

    positioning, several centers use fluoroscopy to check consis-

    tency to the motion state (e.g., exhale) that is being used in

    the treatment planning and (gated) delivery. Fluoroscopy is

    further often used to check the motion range such that the sizeof the aperture is large enough and positioning is accu rate to

    ensure that the tumor moves within the irradiation field.57

    In case of scanned beam delivery, the presence of organ

    motion requires more complex mitigation procedures. Scanned

    beam delivery interferes with target motion typically leading

    to underdosage of the CTV even if margins are used.58,59

    Therefore, currently, only one center treats intrafractionally

    FIG. 2. 4D treatment plan (scanned carbon beam, single field, and absorbeddose) to the GTV of a lung tumor. (a) Planned dose in the reference phase ofthe 4DCT without motion influence. (b) Dose distribution in the presence ofmotion with margins (internal target volume, ITV) as only motion mitiga-tion technique. Interplay of beam scanning and organ motion deteriorateshomogeneity and conformation. (c) Irradiation by beam gating covers the

    target as planned. Data were calculated with the 4D version of GSIs plan-ning code TRiP (Ref. 15).

    1719 Bert, Engenhart-Cabillic, and Durante: Particle therapy for noncancer d iseases 1719

    Medical Physics, Vol. 39, No. 4, April 2012

  • 7/29/2019 Ions for Non Malignancies

    5/12

    moving tumors with a scanned beam using apnea under intu-

    bation and anesthesia.60 Several groups are working on mitiga-

    tion techniques that will allow reliable coverage of the CTV

    also for scanned beams.15 Gating and rescanning will be soon

    translated to clinical usage. Beam tracking is technically feasi-

    ble61,62 but will require robust planning to cover for potential

    changes of the underlying 4DCT data between planning and

    delivery.

    III. DEVELOPMENTS IN THE NEXT DECADE

    III.A. Technical developments

    The cost of particle treatment facilities will decrease in

    the future, both as a result of the interest of large commercial

    companies and of technical developments in the construction

    of compact accelerators, such as fixed alternated gradient

    field syncrocyclotrons,63 ultracompact synchrotrons, and

    laser-driven or dielectric-wall accelerators.64 In the next 10

    years, the number of proton and heavy ion therapy facilitiesworldwide is going to increase rapidly: the reduced cost of

    the accelerator may lead to a reduction in the investmentcost of a factor of 2, but the overall cost of the treatment will

    remain higher than x-ray therapy. The new centers should

    therefore prove an increased benefit (in terms of cure rate

    and/or reduction in overall treatment time) compared to con-

    ventional radiotherapy units. Since high-energy charged par-

    ticles have improved physical characteristics compared to

    x-rays for radiosurgery, all of the current applications of

    SRS or SRT (see Sec. II B) could be gradually tested with

    protons or heavier ions as well. An increase in RBE for the

    therapy-related endpoint might further increase the effective-

    ness for heavier ions compared to photons or protons, as it

    happens in oncology for radioresistant tumors.1 One basic

    problem using heavy ions is the RBE of the particles in the

    different tissues. While the tolerance dose for the various tis-

    sues is known for photons, this is not the case for heavier

    ions. Moreover, the RBE for the target volume can be differ-

    ent from the RBE for the normal tissue surrounding it. This

    problem is the major source of uncertainty in heavy-ion can-

    cer therapy,5 and dedicated research studies are neededbefore extension to new (noncancerous) diseases.

    Apart from these upcoming developments with respect to

    particle type, also an increase in particle energy (non-

    Bragg-peak or plateau particle radiosurgery65) is foreseen

    with applications mainly in SpRS/T. This treatment option

    will not only benefit from reduced lateral beam size but alsoallow online proton radiography exploiting the same thera-

    peutic beam crossing the patient. The combination of both

    physical effects leads to a high precision image-guided ste-

    reotactic particle radiosurgery (IGSpRS).

    III.A.1. New energies

    Proton beam energies used generally in oncology range

    from 60 MeV for uveal melanoma up to 250 MeV for deep

    solid tumors. Higher energies ($GeV) would not allow treat-

    ment of the lesion on the Bragg peak, but radiosurgery will

    still be possible in the plateau region of the Bragg curve

    (Fig. 1), as done with Gammaknife or Cyberknife where the

    depth-dose deposition pattern is opposite to charged par-

    ticles. The rational for using very high-energy protons is

    linked to the reduction in proton scattering.66 Scattering

    causes blurs in the proton treatment plan, which are normally

    less sharp than those obtained with heavier ions. However,

    using protons in the Giga-electron-volt region (b> 0.87), it

    is possible to produce a narrow beam with very sharp edges,

    ideal for ablation of small structures such as AVM. The lat-

    eral scattering is indeed largely reduced compared to Bragg-

    peak protons when relativistic protons are used (Fig. 3). Of

    course, the beam has to be directed from different angles,

    cross-firing the target. As yet, only one facility has used

    1 GeV protons for therapy: the Petersburg Nuclear Physics

    Institute (PNPI) synchrocyclotron in S. Petersburg (Rus-

    sia).67 Since 1975, a total of 1362 patients were treated at

    PNPI for pituitary adenoma, breast and prostate cancer, and

    skull noncancer lesions including AVM, aneurysm, endo-

    crine ophthalmopathy, and epilepsy.

    SRS with relativistic protons (non-Bragg-peak or plateau

    radiosurgery) may be an interesting approach for reducingthe target margins and escalating the target dose. At high

    energy, the beam FWHM will be very narrow (Fig. 4), thus

    ensuring very sharp dose gradients and reduction of the mar-

    gins in the PTV. Although 1 GeV protons have a very low-

    LET ($0.2 keV/lm), they induce several nuclear reactions

    and can produce many neutrons and secondary protons, both

    at high-energy (kick-off protons) and low-energy (evapora-

    tion recoils). These nuclear reactions should be carefully

    considered in the treatment plan, as they can significantly

    modify the physical dose and the biological effectiveness of

    the beam.68 Kick-off protons also broaden the lateral dosedistribution of the beam (Fig. 5), and Monte Carlo codes

    have to be used for accurate predictions (see Figs 4 and 5).Relativistic protons have been extensively studied for space

    radiation protection,69,70 because they represent the major

    component of the galactic cosmic ray flux,71 but very few

    FIG. 3. Simulation using the SRIM2011 code of the spatial distribution ofproton beams (directed to the center) passing through 15 cm of water. Eachdot represents the position of a single proton (scales are in centimeter).Clearly, 1 GeV protons (in plateau) have a much higher spatial resolutionthan beams normally used in therapy (on the Bragg peak).

    1720 Bert, Engenhart-Cabillic, and Durante: Particle therapy for noncancer d iseases 1720

    Medical Physics, Vol. 39, No. 4, April 2012

  • 7/29/2019 Ions for Non Malignancies

    6/12

    studies are available in radiotherapy regime (high dose, bio-

    logical tissues as targets) to be used for in silico trials of

    treatment plans.

    A further advantage of using relativistic protons is that

    the particles traversing the patient can be used for proton ra-

    diography.72 Proton73 and heavy ion74 radiography in medi-

    cine had been proposed many years ago, but the idea wasdropped with the rapid improvements of CT and imaging.

    However, proton radiography is nowadays seriously consid-ered as a sensitive tool for online monitoring in proton ther-

    apy of lung tumors.75 Detection devices include CMOS

    active pixel sensors76 and time-resolved proton range tele-

    scopes.77 Relativistic protons could be used for all kinds of

    lesions in the body, and they have superior spatial and tem-

    poral resolution. About 15 years ago, at Los Alamos

    National Laboratory, it was shown that 800 MeV protons

    could provide improved imaging compared to x-rays of im-

    plosion tests (hydrotest).78 A project for the construction of a

    proton microscope (PRIOR) in the new Facility for Antipro-

    tons and Ion Research (FAIR) in Darmstadt is planning to

    use a 4.5 GeV proton beam able to achieve a resolution

    below 10 lm with a time resolution of 10 ns.79 A combina-

    tion of high-precision radiosurgery with high-resolution radi-

    ography using protons in the Giga-electron-volt energy

    range may represent a new powerful tool for treating several

    types of lesions in the next 10 years. In fact, IGSpRS may

    allow a reduction of target margins and a potential for dose

    escalation. Online imaging opens the possibility of an aim-

    and-shoot technique, and with relativistic protons, the

    imaging reaches unprecedented resolution.

    The different diseases described in the Sec. II B 2 may be

    excellent candidates for SpRS, and especially IGSpRS if

    online radiography is used. As noted in the Sec. II B 2, SRS

    is always considered an attractive alternative to surgery for

    patients nonresponsive to pharmacological treatments,because it is noninvasive and can be repeated especially if

    the dose to OARs is kept low as it would be the case for par-

    ticle beams especially in the Giga-electron-volt regime. One

    of the main problems is the clear delineation of the target,

    especially for epilepsy, and the sparing of structures very

    close to the target, like veins or arteries in trigeminal neural-

    gia, brainstem in epilepsy, and optical nerve and retina in

    macular degeneration. In all these cases, IGSpRS may be

    superior to photon-SRS: using relativistic protons and online

    radiography, the accuracy can go below that achievable in

    SRS, and the target visualization was improved.The cost of the high-energy proton facilities is of course

    very high, and at the moment, it is not realistic to imagineclinical centers; however, trials can be started at several ac-

    celerator facilities already in operation where proton radiog-

    raphy is also implemented, such as FAIR in Darmstadt

    (Germany) and ITEP in Moscow (Russia), or other accelera-

    tor centers with these capabilities such as the Brookhaven

    National Laboratory in USA and CERN in Switzerland. It

    should be noted that particle therapy always started first withtrials in existing nuclear physics facilities, and only in later

    stages, the construction of dedicated centers was planned

    and eventually completed. Moreover, some proton facilities

    using synchrotrons and those using carbon ions (up to 400

    MeV/n) are potentially able to accelerate protons close to 1

    GeV and could then quickly translate in patient treatments in

    the preclinical tests. In silico trials are needed to demonstrate

    that relativistic protons can have an impact in IGRT.

    III.A.2. New ions

    The issue of which particle is optimal for radiotherapy is

    often debated in oncology. Protons are considered safe

    because their RBE is similar to x-rays (a value of 1.1 is used

    in cancer therapy).80 Carbon ions present an optimal plateau/

    peak ratio, with a low-LET in the entrance channel to

    spare the normal tissue, and a relatively high-LET in the

    FIG. 4. Calculations of the beam shapes FWHM for a monoenergetic inci-dent proton beam in water using Molieres formula or simulated by theMonte Carlo code GEANT4. The Moliere theory is an approximation oftenused to calculate the lateral spread of proton beams used in Bragg peak ther-apy (Ref. 5). The FWHM spread is calculated for beams accelerated at60 MeV (used in eyes therapy), 200 MeV (deep protontherapy), 1 GeV(available in several high-energy accelerators, such as the Brookhaven

    National Laboratory in USA and GSI in Germany), 2 and 4.5 GeV (plannedfor proton radiography in the future FAIR facility in Germany). Calculationscourtesy by Dr. Marie Vanstalle (GSI, Germany).

    FIG. 5. Lateral dose distribution of a 1 GeV proton beam passing through a30 mm Al target. The beam was accelerated at the NASA Space RadiationLaboratory (Brookhaven National Laboratory, Upton, NY) and the dosemeasured at different distances from the beam axis using an ionizationchamber (Ref. 68). FWHM of the beam extracted in air was 18 mm. Meas-urements were performed in air at 10 mm from the Al target. Simulations bythe Monte Carlo code PHITS courtesy of Dr. Davide Mancusi (CEA Saclay,France).

    1721 Bert, Engenhart-Cabillic, and Durante: Particle therapy for noncancer d iseases 1721

    Medical Physics, Vol. 39, No. 4, April 2012

  • 7/29/2019 Ions for Non Malignancies

    7/12

    spread-out Bragg-peak (SOBP) to ensure increased RBE and

    reduced oxygen enhancement factor (OER) in the target tu-

    mor. The use of ions with 2 Z 5 can be interesting since

    they have reduced lateral scattering compared to protons but

    lower LET than C-ions. Currently, the Heidelberg Ion Beam

    Therapy Center (HIT) is preparing the installation of a he-

    lium source to allow clinical studies with a helium beam.

    Ions heavier than carbon can be used for very hypoxic

    tumors, when the OER should be reduced more.

    The current investigations toward other projectiles than

    protons and carbon ions in cancer treatment will also apply

    to the choice of optimal ions in noncancer diseases. As

    noted above, He-ions were already used in Berkeley for

    AVM.25,27 In a recent meta-analysis of dose-volume histo-

    grams in treatment of AVM, using a binomial model for

    the number of crucial blood vessels in AVM, Andishehet al.81 concluded that particle radiosurgery generates bet-

    ter dose gradients than x-rays and that for large AVM (>

    10 ml) ions heavier than helium should provide higher

    obliteration rates than light ions. Having the flexibility of

    choosing among different ions sources will greatlyimprove the degrees of freedom of the medical physicists

    in selecting the best possible treatment: lighter ions cover

    the dose more uniformly avoiding any possible cold spot

    in the target, but heavier ions have reduced lateral scatter-

    ing and improved radiobiological properties on the Bragg

    peak.82

    III.B. SBpRT

    With the exception of AVM, only in very few cases that

    charged particles have been used for treating noncancer

    lesions (Sec. II B 1). The new particle therapy centers are,

    however, already considering these pathologies and prepar-ing dedicated treatment rooms. A treatment room completely

    dedicated to radiosurgery has been installed in the newGunma University Heavy Ion Medical Center in Japan,83

    where carbon ions will be used to treat different venous mal-

    formations and macular degeneration in the coming 10 years.

    As noted above, the favorable depth-dose distribution for

    Bragg-peak radiosurgery and the margin reduction expected

    with non-Bragg-peak proton radiosurgery (IGSpRS) may

    provide substantial benefit in several clinical cases. In silico

    trials should be pursued to isolate cases where advantages

    are clear.

    As SBpRT has become a reality in cancer treatment,

    thanks to 4D imaging, improved treatment planning, andimage guidance, SBpRT may also represent a major break-through in radiotherapy of noncancerous lesions in the next

    decade. In the following, we describe the potential of SBpRT

    on the example of atrial fibrillation and renal denervation.

    Since both are subject to organ motion, a dedicated section on

    motion management will follow that is especially required for

    scanned beams that provide maximal conformation.

    III.B.1. Atrial fibrillation

    Cardiac arrhythmia is a common heart disease (preva-

    lence 0.4%1% in the general population with an increase

    by age, leading to 4.5 million patients in the EU),84 which

    can predispose to life-threatening stroke and embolism. The

    treatment of many cardiac arrhythmias is moving from a

    strategy of suppression by antiarrhythmic drugs, which does

    not improve the situation for many patients, to one of poten-

    tial cure by destroying the arrhythmogenic substrate. Abla-

    tion is normally performed by microwaves produced by a

    flexible catheter introduced fluoroscopy guided in a large

    blood vessel and moved in direct contact with the heart

    (catheter ablation).85 While the success rate is >90% for

    some diseases causing arrhythmias, such as the Wolff-Par-

    kinson-White syndrome or the atrial flutter, for atrial fibrilla-

    tion, the success rate of a single catheter ablation drops

    below 30%, requiring multiple invasive interventions.86 This

    seems to be caused by atrial remodeling, especially in

    patients over 50 years old, which makes the heart poorly sus-

    ceptible to ablation. SBRT, as noninvasive method, could

    provide significant advantages.87 A recent experimental

    study in a swine model has proven that Cyberknife (there

    called Cyberheart) can produce cavotricuspid isthmus block,

    AV nodal block, and significant decreased voltage at the pul-monary veinleft atrial junction with no early pathological

    changes in the surrounding tissue.88 However, cavotricuspid

    isthmus block was achieved only in two out of eight animals,

    and the electrophysiological endpoint was delayed and may

    not be clinically relevant.

    These results form the basis for clinical studies on the

    treatment of cardiac arrhythmias with radiotherapy. Can

    charged particles provide better results than the Cyberheart

    in control of atrial fibrillation? Heavy ions may allow a large

    increase in the dose delivered to a small area with sparing of

    the normal tissue. The biological effectiveness of heavy ionsfor the target cells can be high and may overcome resistance

    induced by atrial remodeling and reduce the latency timeobserved with photons. Particle body radiosurgery has there-

    fore the potential of providing an alternative, noninvasive

    management to atrial fibrillation. This hypothesis is under

    study in Japan and Germany (Fig. 6). Nontransmural myo-

    cardial infarction was induced in rabbits by microsphere

    injection into the coronary arteries, and the heart was irradi-

    ated with 15 Gy C-ions 2 weeks after the infarction.89 High-

    energy heavy ions induce upregulation of connexin43

    (Cx43) expression in association with an improvement of

    conduction, a decrease of the spatial inhomogeneity of repo-

    larization, a reduction of vulnerability to ventricular arrhyth-

    mias after myocardial infarction, and no late radiation injury

    up to 1 year after the treatment.90 These results are striking

    because gap junction remodeling creates arrhythmogenic

    substrates by modulating the propagation of excitation, and

    gap junctions in the human heart are mostly made with

    Cx43. Radiation-induced Cx43 upregulation in hearts after

    myocardial infarction can therefore represent an approach in

    the treatment of arrhythmias. A number of experimentalstudies should be completed on the response of cardiac cells

    to high-LET radiation to fully exploit the potential of heavy

    ions and to propose treatment plans using a biological equiv-

    alent ion dose.91 Ion treatment of fibrillations is therefore

    very promising.

    1722 Bert, Engenhart-Cabillic, and Durante: Particle therapy for noncancer d iseases 1722

    Medical Physics, Vol. 39, No. 4, April 2012

  • 7/29/2019 Ions for Non Malignancies

    8/12

    However, for cardiac ablation, it will be necessary to reach

    a 5D treatment, i.e., compensation is necessary for both

    patients breathing and heartbeat: three spatial coordinates

    two time coordinates. This problem will be discussed in the

    Sec. III B 3.

    III.B.2. Renal denervationIn principle, with the treatment of extracranial lesions,

    SBpRT may be competitive to catheter ablation in different

    applications. Another example is renal denervation in

    patients with treatment-resistant hypertension. Severe, resist-

    ant hypertension that is uncontrolled despite patients taking

    antihypertensive medications is a big unmet clinical need,

    with those affected being at increased risk of stroke and renal

    failure. Renal sympathetic efferent and afferent nerves are

    crucial for the initiation and maintenance of systemic hyper-

    tension and lie within and immediately adjacent to the wall

    of the renal artery. The concept of denervation of the renal

    sympathetic nerve to try to reduce blood pressure is old and

    was attempted by surgery some years ago but abandoned

    because of perioperative and long-term side effects.

    Recently, however, it has been shown that renal sympathetic

    nerves can be accessed by a catheter inserted in the femoral

    artery and the nerves ablated by radiofrequency.92 A recentclinical trial has shown that 84% of the patients had a >10

    mm Hg drop in systolic pressure, compared to 34% in the

    control group.93 Kidney function was not altered, and the

    blood pressure reduction is sustained over 2 years from the

    treatment.94 Despite the success of the method, there is a

    16% of nonresponders, without clinical predictors for the

    success. Also, in this case, SBpRT may represent an interest-

    ing alternative. Some patients are not eligible for catheter

    ablation, based on the anatomy of the kidney arteries (exam-

    ined by angiography). Transcatheters target 45 sites at each

    renal sympathetic nerve, which would be the targets for

    radiosurgery. The dose necessary for the radiotherapeutical

    denervation is unknown. The experience with stereotactic

    radiotherapy in trigeminal neuralgia (Sec. II B 2) can be

    translated to the sympathetic nerve. Further indications onthe peripheral nerve tolerance doses can be obtained by the

    incidence of brachial plexopathy95 following radiotherapy,especially for breast cancer. Plexopathy is generally a late

    effect, which is observed after a few months from the treat-

    ment. The tolerance dose is about 60 Gy, but it is markedly

    reduced by hypofractionation,96 suggesting that hypofractio-

    nated SBpRT can be exquisitely effective in denervation.

    Treatment of the kidney requires careful management of the

    respiratory motion: motion amplitudes up to 40 mm have

    been reported.97 Detailed thoughts are given in Sec. III B 3.

    III.B.3. 4D- and 5D-treatment plans for particle bodyradiotherapy

    The predominant cause of motion in SBpRT is respiration

    that affects both of the chosen example sites, i.e., heart and

    kidney. Management of respiratory motion in scanned parti-cle beam therapy including the vision for the next years have

    previously been addressed with respect to cancer ther-

    apy.15,98 Treatment of patients with respiration influences

    tumors has already been started for proton as well as carbon

    beam therapy using abdominal compression (HIT, Germany)

    and apnea (RPTC Munich).60 Treatment with gating at HIT

    can be expected by early 2012 and NIRS as well as PSI will

    FIG. 6. IMRT plan for treatment of atrial fibrillation by photon beam therapy produced within a feasibility planning study. (a) and (b) show each seven copla-nar beams that are used to treat the left pulmonary vein (LPV) and the right pulmonary vein (RPV), respectively. Apart from the beam portals and the target

    contour each figure contains isodose lines for 9 Gy (light blue), 15 Gy (magenta), and 25 Gy (yellow) and the contours of various organs at risk [OAR, bron-chial tree in blue, esophagus in green, spinal cord in light blue, left coronary artery (LCA) in magenta]. A 3D overview of the irradiation geometry is providedin (c) with both target volumes indicated in red, the 25 Gy isodose in yellow, and in OARs in the colors indicated above. Dose-volume histograms are shownin (d) for IMRT (~) and 3D conformal (n) treatment delivery options. Images courtesy of Dr. O. Blanck (UKSH Lubeck, Germany).

    1723 Bert, Engenhart-Cabillic, and Durante: Particle therapy for noncancer d iseases 1723

    Medical Physics, Vol. 39, No. 4, April 2012

  • 7/29/2019 Ions for Non Malignancies

    9/12

    treat tumors with rescanning (in combination with gating atNIRS) in the next years.

    The techniques used for treatment of moving tumors will

    also be appropriate to treat noncancerous lesions like renaldenervation. In this specific example, the most natural way

    to irradiate the kidney, especially for a low number of beam

    positions as required for ablation treatment of renal denerva-

    tion, should be a gated irradiation as shown in Fig. 2 for a

    lung tumor.99 Motion monitoring could either be achieved

    by one of the established motion detection systems (e.g.,

    ANZAI belt, VARIAN RPM, and Cyberknife Synchrony

    correlation model) but kidney motion is also detectable via

    ultrasound,97 which would provide internal motion ratherthan a surrogate. NIRS in Japan performs treatment of renal

    cell carcinoma with a passively scattered carbon beam since

    1997.100 They use gating based on the skin motion as surro-

    gate to suppress the impact of respiratory motion. Patientpositioning is performed via fluoroscopy using implanted

    iridium needles.

    For other sites rescanning, beam tracking or combinations

    motion mitigation techniques will be appropriate. In general,

    also for noncancer lesions, the concluding remarks of Rietzel

    and Bert98 are valid, i.e., patients will be classified according

    to their motion characteristics and treated with a single or

    combinations of the established motion mitigation techniques.

    For treatment of cardiac lesions, respiration and heart

    beat have to be compensated. In the Cyberheart study in

    swines,88 beam tracking was used to compensate respiratory

    motion. Heart motion was incorporated by margins. This

    mode of operation is in principle available today for all treat-

    ment options that can cope with respiratory motion, includ-

    ing heavy-ion therapy that is indeed used to treat cardiac

    angiosarcomas.101 In order to exploit the full potential of

    SBpRT, more conformal treatment options would be pre-ferred. This is possible by considering the motion of heart

    and respiration independently (Fig. 7) resulting into a 5D

    treatment scheme. For each of the two motions, one of the

    previously mentioned techniques can be used resulting in

    treatment combinations such as respiratory gating combined

    with cardiac gating or double beam tracking based on car-

    diac as well as respiratory motion. However, specific

    improvements are needed to address the 5D treatment work-flow. Changes are not only required in the treatment delivery

    modules but also toward imaging, treatment planning,

    patient positioning, and quality assurance. Details will oncemore depend on the specific site and the facility used for

    treatment delivery. For atrial fibrillation as the example,

    catheter ablation requires a few hundred ablation points,

    which should correspond to one Bragg-peak position each.

    In cyclotron based treatments with passive energy degrada-

    tion like at PSI, scanning of 100 points would take less than

    10 s even if each point requires a different energy since

    energy changes are feasibly in 80 ms.102 Treatments could

    then be delivered based on a respiratory gated cardiac 4DCTwith the appropriate deformation maps and 4D (cardiac)

    treatment planning for beam tracking resulting in transfor-

    mation parameters for each beam position with respect to the

    cardiac reference phase.103,104 Motion surrogates for beamdelivery would be identical to CT imaging, e.g., chest wall

    motion plus ECG triggering. These systems could also be

    used for cardiac respiratory gated setup using orthogonal

    x-ray imaging of a potentially clipped target. Quality assur-

    ance will be an important but challenging topic. But also the

    results and methods that are currently translated into clinical

    use for 4D therapy of respiratory moving tumors can be

    used. For example, various groups develop motion phantoms

    that can be used for plan verification 105107 but also 4DPET

    can be an option for proton as well as carbon beams. Initial

    phantom studies were successful,108 and translation to clini-

    cal routine is currently performed for treatment of hepatocel-

    lular cancer patients at HIT.

    IV. CONCLUSIONS

    In the coming 10 years, particle therapy centers should beable to treat several noncancer diseases and to provide an

    effective stereotactic radiosurgery, provided that research

    studies ongoing and planned will give positive results.

    Research accelerator facilities should be used for testing the

    applications of new ions and very high-energy ($GeV) pro-

    tons, also very promising for their potential in simultaneous

    radiography (image-guided stereotactic particle radiosurgery).

    FIG. 7. The combined 5D motion of heart beat and res-piration requires different treatment concepts. In princi-ple, for each of the two motion dimensions, one of themotion mitigation techniques that were proposed for re-

    spiratory motion (margins, rescanning, gating, beamtracking) has to be used. Depending on the chosencombination treatment efficiency and target volumeconformation will change.

    1724 Bert, Engenhart-Cabillic, and Durante: Particle therapy for noncancer d iseases 1724

    Medical Physics, Vol. 39, No. 4, April 2012

  • 7/29/2019 Ions for Non Malignancies

    10/12

    Particle body radiosurgery may have breakthrough applica-

    tions where currently invasive catheter ablation techniques

    are needed, e.g., atrial fibrillation or renal sympathetic dener-

    vation. A 5D treatment planning system should be developed

    and tested in animal models. The RBE of the charged particles

    must be studied in the tissue of interest at high doses. Options

    for Bragg-peak or relativistic plateau proton-radiosurgery

    should be considered in the debate about cost/benefit ratio

    currently ongoing about the construction of new accelerator-

    based therapy facilities. In silico trials (including Bragg-peak

    protons, plateau protons, heavier ions with 2 Z 8, and of

    course x-rays orc-rays) should be pursued for these different

    diseases to highlight possible cases where charged particles

    may provide a clear clinical advantage. Preclinical radiobiol-

    ogy studies should then corroborate the treatment plan predic-

    tions, and finally clinical trials can be planned.

    ACKNOWLEDGMENT

    Research on particle therapy at GSI is partly supported by

    SIEMENS AG and EU FP7 project ULICE and ENVISION.

    a)Electronic address: [email protected]. Durante and J. S. Loeffler, Charged particles in radiation oncology,

    Nat. Rev. Clin. Oncol. 7, 3743 (2010).2A. R. Smith, Vision 20/20: proton therapy, Med. Phys. 36, 556568

    (2009).3O. Jakel, C. P. Karger, and J. Debus, The future of heavy ion radio-

    therapy, Med. Phys. 35, 56535663 (2008).4D. Schulz-Ertner and H. Tsujii, Particle radiation therapy using proton

    and heavier ion beams, J. Clin. Oncol. 25, 953964 (2007).5D. Schardt, T. Elsasser, and D. Schulz-Ertner, Heavy-ion tumor therapy:

    Physical and radiobiological benefits, Rev. Mod. Phys. 82, 383425(2010).

    6

    T. E. Merchant, Proton beam therapy in pediatric oncology, Cancer J.15, 298305 (2009).7W. D. Newhauser, and M. Durante, Assessing the risk of second malig-

    nancies after modern radiotherapy, Nat. Rev. Cancer 11, 438448(2011).

    8T. Terasawa, T. Dvorak, S. Ip, G. Raman, J. Lau, and T. A. Trikalinos,Systematic review: Charged particle therapy for cancer, Ann. Intern.Med. 151, 556565 (2009).

    9E. C. Halperin, Particle therapy and treatment of cancer, Lancet Oncol.7, 676685 (2006).

    10Particle Therapy Co-Operative group homepage. http://ptcog.web.psi.ch11P. Pommier, Y. Lievens, F. Feschet, J. M. Borras, M. H. Baron, A. Shti-

    liyanova, and M. Pijls-Johannesma, Simulating demand for innovativeradiotherapies: An illustrative model based on carbon ion and protonradiotherapy, Radiother. Oncol. 96, 243249 (2010).

    12M. Brada, M. Pijls-Johannesma, and D. De Ruysscher, Current clinical

    evidence for proton therapy, Cancer J.15

    , 319324 (2009).13D. Benedict, F. J. Bova, B. Clark, S. J. Goetsch, W. H. Hinson, D. D.Leavitt, D. J. Schlesinger, and K. M. Yenice, Anniversary paper: Therole of medical physicists in developing stereotactic radiation therapy,Med. Phys. 35, 42624277 (2008).

    14S. Dietrich, K. Cleary, W. DSouza, M. Murphy, K. H. Wong, and P.Keall, Locating and targeting moving tumors with radiation beams,Med. Phys. 35, 56845691 (2008).

    15C. Bert, and M. Durante, Motion in radiotherapy: particle therapy,Phys. Med. Biol. 56, R113R144 (2011).

    16H. Suit, H. Krooy, A. Trofimov, J. Farr, J. Munzenrider, T. DeLaney,J. S. Loeffler, B. Clasie, S. Safai, and H. Paganetti, Should positivephase III clinical trial data be required before proton beam therapy ismore widely adopted? No, Radiother. Oncol. 86, 148153 (2008).

    17H. Suit, T. DeLaney, S. Goldberg, H. Paganetti, B. Clasie, L. Germeck,A. Niemerko, E. Hall, J. Flanz, J. Hallman, and A. Trofimov, Proton vs.

    carbon ion beams in the definitive radiation treatment of cancer patients,Radiother. Oncol. 95, 322 (2010).

    18B. Larsson, L. Leksell, B. Rexed, P. Sourander, W. Mair, and B. Ander-sson, The high-energy proton beam as a neurosurgical tool, Nature 182,12221223 (1958).

    19L. Leksell, Radiosurgery, Neurosurgery 24, 297298 (1989).20J. D. Fenwick, W. A. Tome, E. T. Soisson, M. P. Mehta, and T. Rock

    Mackie, Tomotherapy and other innovative IMRT delivery systems,Semin. Radiat. Oncol. 16, 199208 (2006).

    21R. N. Kjellberg, T. Hanamura, K. R. Davis, S. L. Lyons, and R. D.Adams, Bragg-peak proton-beam therapy for arteriovenous malforma-tions of the brain, N. Engl. J. Med. 30, 269274 (1983).

    22V. Seifert, D. Stolke, H. M. Mehdorn, and B. Hoffmann, Clinical and ra-diological evaluation of long-term results of stereotactic proton beamradiosurgery in patients with cerebral arteriovenous malformations,J. Neurosurg. 81, 683689 (1994).

    23F. G. Barker II, W. E. Butler, S. Lyons, E. Cascio, C. S. Ogilvy,J. S. Loeffler, and P. H. Chapman, Dose-volume prediction ofradiation-related complications after proton beam radiosurgery forcerebral arteriovenous malformations, J. Neurosurg. 99, 254263(2003).

    24C. C. Chen, P. Chapman, J. Petit, and J. S. Loeffler, Proton radiosurgeryin neurosurgery, Neurosurg. Focus 23, E5 (2007).

    25R. P. Levy, J. I. Fabrikant, K. A. Frankel, M. H. Phillips, and J. T.Lyman, Stereotactic heavy-charged-particle Bragg peak radiosurgery forthe treatment of intracranial arteriovenous malformations in childhoodand adolescence, Neurosurgery 24, 841852 (1989).

    26H. Silander, L. Pellettieri, P. Enblad, A. Montelius, E. Grusell, C.Vallhagen-Dahlgren, U. Isacsson, G. Nyberg, U. Mostrom, A. Lilja, G.Gal, and E. Blomquist, Fractionated, stereotactic proton beam treatmentof cerebral arteriovenous malformations, Acta Neurol. Scand. 109,8590 (2004).

    27G. K. Steinberg, J. I. Fabrikant, M. P. Marks, R. P. Levy, K. A. Frankel,M. H. Phillips, L. M. Shuer, and G. D. Silverberg, Stereotactic heavy-charged-particle Bragg-peak radiation for intracranial arteriovenousmalformations, N. Engl. J. Med. 323, 96101 (1990).

    28F. J. A. I. Vernimmen, J. P. Slabbert, J. A. Wilson, S. Fredericks, andR. Melvill, Stereotactic proton beam therapy for intracranial arterove-nous malformations, Int. J. Radiat. Oncol., Biol., Phys. 62, 4452(2005).

    29B. C. Lopez, P. J. Hamlyn, and J. M. Zakrzewska, Stereotactic radiosur-gery for primary trigeminal neuralgia: State of the evidence and recom-mendations for future reports, J. Neurol. Neurosurg. Psychiatry 75,10191024 (2004).

    30M. Quigg and N. M. Barbaro, Stereotactic radiosurgery for treatment ofepilepsy, Arch Neurol. 65, 177183 (2008).

    31B. W. Chong, Current issues in endovascular surgical neuroradiology,Semin. Neurol. 27, 385392 (2007).

    32J. Hanlon, C. Lee, E. Chell, M. Gertner, S. Hansen, R. W. Howell, andW. E. Bolch, Kilovoltage stereotactic radiosurgery for age-related macu-lar degeneration: Assessment of optic nerve dose and patient effectivedose, Med. Phys. 36, 36713681 (2009).

    33K. Tipton, J. H. Launders, R. Inamdar, C. Miyamoto, and K. Schoelles,Stereotactic body radiation therapy: Scope of the literature, Ann. In-tern. Med. 154, 737745 (2011).

    34J. M. Zakrzewska and M. E. Linskey, Trigeminal neuralgia, Clin. Evid.12, 1207 (2009).

    35G. C. Jones, A. L. Elaimy, J. J. Demakas, H. Jiang, W. T. Lamoreaux, R.K. Fairbanks, A. R. Mackay, B. S. Cooke, and C. M. Lee, Feasibility ofmultiple repeat gamma knife radiosurgeries for trigeminal neuralgia: Acase report and review of the literature, Case Report Med. 2011, 258910(2011).

    36A. C. Aubuchon, M. D. Chan, J. F. Lovato, C. J. Balamucki, T. L. Ellis,S. B. Tatter, K. P. McMullen, M. T. Munley, A. F. Deguzman, K. E.Ekstrand, J. D. Bourland, and E. G. Shaw, Repeat gamma knife radio-surgery for trigeminal neuralgia, Int. J. Radiat. Oncol., Biol., Phys. 81,10591065 (2011).

    37T. H. Schwarz, Predicting the unpredictable: Stereotactic radiosurgeryand temporal lobe epilepsy, Epilepsy Curr. 10, 150152 (2010).

    38E. F. Chang, M. Quigg, M. C. Oh, W. P. Dillon, M. M. Ward, K. D.Laxer, D. K. Broshek, and N. M. Barbaro; Epilepsy Radiosurgery StudyGroup, Predictors of efficacy after stereotactic radiosurgery for medialtemporal lobe epilepsy, Neurology 74, 165172 (2010).

    1725 Bert, Engenhart-Cabillic, and Durante: Particle therapy for noncancer d iseases 1725

    Medical Physics, Vol. 39, No. 4, April 2012

    http://dx.doi.org/10.1038/nrclinonc.2009.183http://dx.doi.org/10.1118/1.3058485http://dx.doi.org/10.1118/1.3002307http://dx.doi.org/10.1200/JCO.2006.09.7816http://dx.doi.org/10.1103/RevModPhys.82.383http://dx.doi.org/10.1097/PPO.0b013e3181b6d4b7http://dx.doi.org/10.1038/nrc3069http://dx.doi.org/10.1016/S1470-2045(06)70795-1http://ptcog.web.psi.ch/http://dx.doi.org/10.1016/j.radonc.2010.04.010http://dx.doi.org/10.1097/PPO.0b013e3181b6127chttp://dx.doi.org/10.1118/1.2969268http://dx.doi.org/10.1118/1.3020593http://dx.doi.org/10.1088/0031-9155/56/16/R01http://dx.doi.org/10.1016/j.radonc.2007.12.024http://dx.doi.org/10.1016/j.radonc.2010.01.015http://dx.doi.org/10.1038/1821222a0http://dx.doi.org/10.1097/00006123-198902000-00026http://dx.doi.org/10.1016/j.semradonc.2006.04.002http://dx.doi.org/10.1056/NEJM198308043090503http://dx.doi.org/10.3171/jns.1994.81.5.0683http://dx.doi.org/10.3171/jns.2003.99.2.0254http://dx.doi.org/10.3171/FOC-07/10/EIntrohttp://dx.doi.org/10.1227/00006123-198906000-00009http://dx.doi.org/10.1046/j.1600-0404.2003.00154.xhttp://dx.doi.org/10.1056/NEJM199007123230205http://dx.doi.org/10.1016/j.ijrobp.2004.09.008http://dx.doi.org/10.1136/jnnp.2003.018564http://dx.doi.org/10.1001/archneurol.2007.40http://dx.doi.org/10.1055/s-2007-985339http://dx.doi.org/10.1118/1.3168554http://dx.doi.org/10.1016/j.ijrobp.2010.07.010http://dx.doi.org/10.1111/epc.2010.10.issue-6http://dx.doi.org/10.1212/WNL.0b013e3181c9185dhttp://dx.doi.org/10.1212/WNL.0b013e3181c9185dhttp://dx.doi.org/10.1111/epc.2010.10.issue-6http://dx.doi.org/10.1016/j.ijrobp.2010.07.010http://dx.doi.org/10.1118/1.3168554http://dx.doi.org/10.1055/s-2007-985339http://dx.doi.org/10.1001/archneurol.2007.40http://dx.doi.org/10.1136/jnnp.2003.018564http://dx.doi.org/10.1016/j.ijrobp.2004.09.008http://dx.doi.org/10.1056/NEJM199007123230205http://dx.doi.org/10.1046/j.1600-0404.2003.00154.xhttp://dx.doi.org/10.1227/00006123-198906000-00009http://dx.doi.org/10.3171/FOC-07/10/EIntrohttp://dx.doi.org/10.3171/jns.2003.99.2.0254http://dx.doi.org/10.3171/jns.1994.81.5.0683http://dx.doi.org/10.1056/NEJM198308043090503http://dx.doi.org/10.1016/j.semradonc.2006.04.002http://dx.doi.org/10.1097/00006123-198902000-00026http://dx.doi.org/10.1038/1821222a0http://dx.doi.org/10.1016/j.radonc.2010.01.015http://dx.doi.org/10.1016/j.radonc.2007.12.024http://dx.doi.org/10.1088/0031-9155/56/16/R01http://dx.doi.org/10.1118/1.3020593http://dx.doi.org/10.1118/1.2969268http://dx.doi.org/10.1097/PPO.0b013e3181b6127chttp://dx.doi.org/10.1016/j.radonc.2010.04.010http://ptcog.web.psi.ch/http://dx.doi.org/10.1016/S1470-2045(06)70795-1http://dx.doi.org/10.1038/nrc3069http://dx.doi.org/10.1097/PPO.0b013e3181b6d4b7http://dx.doi.org/10.1103/RevModPhys.82.383http://dx.doi.org/10.1200/JCO.2006.09.7816http://dx.doi.org/10.1118/1.3002307http://dx.doi.org/10.1118/1.3058485http://dx.doi.org/10.1038/nrclinonc.2009.183
  • 7/29/2019 Ions for Non Malignancies

    11/12

    39A. Yuan and P. K. Kaiser, Emerging therapies for the treatment of neo-vascular age related macular degeneration, Semin. Ophthalmol. 26,149155 (2011).

    40R. Petrarca and T. L. Jackson, Radiation therapy for neovascular age-related macular degeneration, Clin. Ophthalmol. 5, 5763 (2011).

    41M. Gertner, E. Chell, K. H. Pan, S. Hansen, P. K. Kaiser, and D. M.Moshfeghi, Stereotactic targeting and dose verification for age-relatedmacular degeneration, Med. Phys. 37, 600606 (2010).

    42H. J. Zambarakji, A. M. Lane, E. Ezra, D. Gauthier, M. Goitein, J. A.Adams, J. E. Munzenrider, J. W. Miller, and E. S. Gragoudas, Protonbeam irradiation for neovascular age-related macular degeneration, Oph-thalmology 113, 20122019 (2006).

    43S. M. Hahn and A. Maity, General principles of radiation and chemo-radiation, Retina 29, S30S31 (2009).

    44K. V. Chalam, S. Balaiya, R. S. Malyappa, W. Hsi, V. S. Brar, and R. K.Murthy, Evaluation of choroidal endothelial cell proliferation after ex-posure to varying doses of proton beam radiation, Retina 31, 169176(2011)

    45T. Miyamoto, M. Baba, T. Sugane, M. Nakajima, T. Yashiro, K. Kagei,N. Hirasawa, T. Sugawara, N. Yamamoto, M. Koto, H. Ezawa, K.Kadono, H. Tsujii, J. E. Mizoe, K. Yoshikawa, S. Kandatsu, and T. Fuji-sawa; Working Group for Lung Cancer, Carbon ion radiotherapy forstage I non-small cell lung cancer using a regimen of four fractions dur-ing 1 week, J. Thorac. Oncol. 2, 916926 (2007).

    46Z. Liao, S. H. Lin, and J. D. Cox, Status of particle therapy for lung can-cer, Acta Oncol. 50, 745756 (2011).

    47T. Chiba, K. Tokuuye, Y. Matsuzaki, S. Sugahara, Y. Chuganji, K. Kagei,J. Shoda, M. Hata, M. Abei, H. Igaki, N. Tanaka, and Y. Akine, Protonbeam therapy for hepatocellular carcinoma: A retrospective review of162 patients, Clin. Cancer Res. 11, 37993805 (2005).

    48H. Kato, H. Tsujii, T. Miyamoto, J. E. Mizoe, T. Kamada, H. Tsuji, S.Yamada, S. Kandatsu, K. Yoshikawa, T. Obata, H. Ezawa, S. Morita, M.Tomizawa, N. Morimoto, J. Fujita, and M. Ohto; Liver Cancer WorkingGroup, Results of the first prospective study of carbon ion radiotherapyfor hepatocellular carcinoma with liver cirrhosis, Int. J. Radiat. Oncol.,Biol., Phys. 59, 14681476 (2004).

    49T. Okada, T. Kamada, H. Tsuji, J. E. Mizoe, M. Baba, S. Kato, S.Yamada, S. Sugahara, S. Yasuda, N. Yamamoto, R. Imai, A. Hasegawa,H. Imada, H. Kiyohara, K. Jingu, M. Shinoto, and H. Tsujii, Carbon ionradiotherapy: Clinical experiences at National Institute of RadiologicalScience (NIRS), J. Radiat. Res. 51, 355364 (2010).

    50S. Mori, G. T. Chen, and M. Endo, Effects of intrafractional motion onwater equivalent path length in respiratory gated heavy charged particlebeam radiotherapy, Int. J. Radiat. Oncol., Biol., Phys. 69, 308317(2007).

    51H. Tsuji, T. Yanagi, H. Ishikawa, T. Kamada, J. E. Mizoe, T. Kanai, S.Morita, and H. Tsujii; Working Group for Genitourinary Tumors,Hypofractionated radiotherapy with carbon ion beams for prostate can-cer, Int. J. Radiat. Oncol., Biol., Phys. 63, 11531160 (2005).

    52D. A. Bush, J. D. Slater, B. B. Shin, G. Cheek, D. W. Miller, and J. M.Slater, Hypofractionated proton beam radiotherapy for stage I lung can-cer, Chest 126, 11981203 (2004).

    53T. Miyamoto, N. Yamamoto, H. Nishimura, M. Koto, H. Tsujii, J. E.Mizoe, T. Kamada, H. Kato, S. Yamada, S. Morita, K. Yoshikawa, S.Kandatsu, and T. Fujisawa, Carbon ion radiotherapy for stage I non-small cell lung cancer, Radiother. Oncol. 66, 127140 (2003).

    54R. J. Smeenk, B. S. Teh, E. B. Butler, E. N. van Lin, and J. H. Kaanders,Is there a role for endorectal balloons in prostate radiotherapy? A sys-tematic review, Radiother. Oncol. 95, 277282 (2010).

    55M. Urie, M. Goitein, and M. Wagner, Compensating for heterogeneitiesin proton radiation therapy, Phys. Med. Biol. 29, 553566 (1984).

    56M. Koto, T. Miyamoto, N. Yamamoto, H. Nishimura, S. Yamada, and H.Tsujii, Local control and recurrence of stage I non-small cell lung cancerafter carbon ion radiotherapy, Radiother. Oncol. 71, 147156 (2004).

    57S. Minohara, T. Kanai, M. Endo, K. Noda, and M. Kanazawa,Respiratory gated irradiation system for heavy-ion radiotherapy, Int. J.Radiat. Oncol., Biol., Phys. 47, 10971103 (2000).

    58M. H. Phillips, E. Pedroni, H. Blattmann, T. Boehringer, A. Coray, and S.Scheib, Effects of respiratory motion on dose uniformity with a chargedparticle scanning method, Phys. Med. Biol. 37, 223233 (1992).

    59C. Bert, S. O. Grozinger, and E. Rietzel, Quantification of interplayeffects of scanned particle beams and moving targets, Phys. Med. Biol.53, 22532265 (2008).

    60M. Eckermann, Scanning proton beam radiotherapy under functionalapnea, PTCOG 50 2011 (presentation and abstract).

    61C. Bert, A. Gemmel, N. Saito, N. Chaudhri, D. Schardt, M. Durante, G.Kraft, and E. Rietzel, Dosimetric precision of an ion beam trackingsystem, Radiat. Oncol. 5, 61 (2010).

    62N. Saito, C. Bert, N. Chaudhri, A. Gemmel, D. Schardt, M. Durante, andE. Rietzel, Speed and accuracy of a beam tracking system for treatmentof moving targets with scanned ion beams, Phys. Med. Biol. 54,48494862 (2009).

    63D. Clery, The next big beam?, Science 327, 142143 (2010).64G. Kraft and S. D. Kraft, Research needed for improving heavy-ion

    therapy, New J. Phys. 11, 025001 (2009).65S. M. Vatnitsky, D. W. Miller, M. F. Moyers, R. P. Levy, R. W. Schulte,

    J. D. Slater, and J. M. Slater, Dosimetry techniques for narrow protonbeam radiosurgery, Phys. Med. Biol. 44, 27892801 (1999).

    66J. M. Schippers and A. J. Lomax, Emerging technologies in protontherapy, Acta Oncol. 50, 838850 (2011).

    67N. K. Abrosimov, Y. A. Gavrikov, E. M. Ivanov, D. L. Karlin, A. V.Khanzadeev, N. N. Yalynych, G. A. Riabov, D. M. Seliverstov, and V.M. Vinogradov, 1000 MeV proton therapy facility at Petersburg NuclearPhysics Institute Synchrocyclotron, J. Phys. 41, 424432 (2006).

    68A. Bertucci, M. Durante, G. Gialanella, G. F. Grossi, L. Manti, M. Pugli-ese, P. Scampoli, D. Mancusi, L. Sihver, and A. Rusek A., Shielding ofrelativistic protons, Radiat. Environ. Biophys. 46, 107111 (2007).

    69H. Yang, N. Magpayo, and K. D. Held, Targeted and non-targetedeffects from combination of low doses of energetic protons and iron ionsin human fibroblasts, Int. J. Radiat. Biol. 87, 311319 (2011).

    70M. Durante and F. A. Cucinotta, Heavy ion carcinogenesis and humanspace exploration, Nat. Rev. Cancer8, 465472 (2008).

    71M. Durante and F. A. Cucinotta, Physical basis of radiation protection inspace travel, Rev. Mod. Phys. 83, 12451281 (2011).

    72A. Niranjan and J. C. Flickinger, Radiobiology, principle and techniqueof radiosurgery, Prog. Neurol. Surg. 21, 3242 (2008).

    73A. M. Koehler, Proton radiography, Science 160, 303 (1968).74C. A. Tobias, The future of heavy-ion science in biology and medicine,

    Radiat. Res. 103, 133 (1985).75N. Depauw and J. Seco, Sensitivity study of proton radiography and

    comparison with kV and MV x-ray imaging using GEANT4 Monte Carlosimulations, Phys. Med. Biol. 56, 24072421 (2011).

    76J. Seco and N. Depauw, Proof of principle study of the use of a CMOSactive pixel sensor for proton radiography, Med. Phys. 38, 622623(2011).

    77B. Han, X. G. Xu, and G. T. Chen, Proton radiography and fluoroscopyof lung tumors: A Monte Carlo study using patient-specific 4DCTphantoms, Med. Phys. 38, 19031911 (2011).

    78P. A. Rigg, C. L. Schwartz, R. S. Hixson, G. E. Hogan, K. K. Kwiatkow-ski, F. G. Mariam, M. Marr-Lyon, F. E. Merrill, C. L. Morris, P. Rightly,A. Saunders, and D. Tupa, Proton radiography and accurate densitymeasurements: A window into shock wave processes, Phys. Rev. B 77,220101 (2008).

    79F. E. Merrill, A. A. Golubev, F. G. Mariam, V. I. Turtikov, and D. Var-entsov, Proton microscopy at FAIR, AIP Conf. Proc. 1195, 667670(2009).

    80H. Paganetti, Interpretation of proton relative biological effectivenessusing lesion induction, lesion repair, and cellular dose distribution, Med.Phys. 32, 25482556 (2005).

    81B. Andisheh, A. Brahme, M. A. Bitaraf, P. Mavroidis, and B. K. Lind,Clinical and radiobiological advantages of single-dose stereotactic light-ion radiation therapy for large intracranial arteriovenous malformations.Technical note, J. Neurosurg. 111, 919926 (2009).

    82A. Brahme, Recent advances in light ion therapy, Int. J. Radiat. Oncol.Biol. Phys. 58, 203216 (2004).

    83Gunma University Heavy Ion Medical Center webpage. http://heavy-ion.showa.gunma-u.ac.jp/en/index.html

    84V. Fuster, L. E. Ryden, D. S. Cannom, H. J. Crijns, A. B. Curtis, K. A.Ellenbogen, J. L. Halperin, J. Y. Le Heuzey, G. N. Kay, J. E. Lowe, S. B.Olsson, E. N. Prystowsky, J. L. Tamargo, S. Wann, S. C. Smith, Jr., A. K.Jacobs, C. D. Adams, J. L. Anderson, E. M. Antman, J. L. Halperin, S. A.Hunt, R. Nishimura, J. P. Ornato, R. L. Page, B. Riegel, S. G. Priori, J. J.Blanc, A. Budaj, A. J. Camm, V. Dean, J. W. Deckers, C. Despres, K.Dickstein, J. Lekakis, K. McGregor, M. Metra, J. Morais, A. Osterspey, J.L. Tamargo, and J. L. Zamorano; American College of Cardiology,American Heart Association Task Force, European Society of Cardiology

    1726 Bert, Engenhart-Cabillic, and Durante: Particle therapy for noncancer d iseases 1726

    Medical Physics, Vol. 39, No. 4, April 2012

    http://dx.doi.org/10.3109/08820538.2011.570846http://dx.doi.org/10.2147/OPTHhttp://dx.doi.org/10.1118/1.3291648http://dx.doi.org/10.1016/j.ophtha.2006.05.036http://dx.doi.org/10.1016/j.ophtha.2006.05.036http://dx.doi.org/10.1097/IAE.0b013e3181ad2692http://dx.doi.org/10.1097/IAE.0b013e3181dee621http://dx.doi.org/10.1097/JTO.0b013e3181560a68http://dx.doi.org/10.3109/0284186X.2011.590148http://dx.doi.org/10.1158/1078-0432.CCR-04-1350http://dx.doi.org/10.1016/j.ijrobp.2004.01.032http://dx.doi.org/10.1016/j.ijrobp.2004.01.032http://dx.doi.org/10.1269/jrr.10016http://dx.doi.org/10.1016/j.ijrobp.2007.05.018http://dx.doi.org/10.1016/j.ijrobp.2005.04.022http://dx.doi.org/10.1378/chest.126.4.1198http://dx.doi.org/10.1016/S0167-8140(02)00367-5http://dx.doi.org/10.1016/j.radonc.2010.04.016http://dx.doi.org/10.1088/0031-9155/29/5/008http://dx.doi.org/10.1016/j.radonc.2004.02.007http://dx.doi.org/10.1016/S0360-3016(00)00524-1http://dx.doi.org/10.1016/S0360-3016(00)00524-1http://dx.doi.org/10.1088/0031-9155/37/1/016http://dx.doi.org/10.1088/0031-9155/53/9/003http://dx.doi.org/10.1186/1748-717X-5-61http://dx.doi.org/10.1088/0031-9155/54/16/001http://dx.doi.org/10.1126/science.327.5962.142http://dx.doi.org/10.1088/1367-2630/11/2/025001http://dx.doi.org/10.1088/0031-9155/44/11/308http://dx.doi.org/10.3109/0284186X.2011.582513http://dx.doi.org/10.1088/1742-6596/41/1/047http://dx.doi.org/10.1007/s00411-006-0088-6http://dx.doi.org/10.3109/09553002.2010.537431http://dx.doi.org/10.1038/nrc2391http://dx.doi.org/10.1103/RevModPhys.83.1245http://dx.doi.org/10.1159/000156557http://dx.doi.org/10.1126/science.160.3825.303http://dx.doi.org/10.2307/3576668http://dx.doi.org/10.1088/0031-9155/56/8/006http://dx.doi.org/10.1118/1.3496327http://dx.doi.org/10.1118/1.3555039http://dx.doi.org/10.1103/PhysRevB.77.220101http://dx.doi.org/10.1063/1.3295228http://dx.doi.org/10.1118/1.1949807http://dx.doi.org/10.1118/1.1949807http://dx.doi.org/10.3171/2007.10.17205http://heavy-ion.showa.gunma-u.ac.jp/en/index.htmlhttp://heavy-ion.showa.gunma-u.ac.jp/en/index.htmlhttp://heavy-ion.showa.gunma-u.ac.jp/en/index.htmlhttp://heavy-ion.showa.gunma-u.ac.jp/en/index.htmlhttp://dx.doi.org/10.3171/2007.10.17205http://dx.doi.org/10.1118/1.1949807http://dx.doi.org/10.1118/1.1949807http://dx.doi.org/10.1063/1.3295228http://dx.doi.org/10.1103/PhysRevB.77.220101http://dx.doi.org/10.1118/1.3555039http://dx.doi.org/10.1118/1.3496327http://dx.doi.org/10.1088/0031-9155/56/8/006http://dx.doi.org/10.2307/3576668http://dx.doi.org/10.1126/science.160.3825.303http://dx.doi.org/10.1159/000156557http://dx.doi.org/10.1103/RevModPhys.83.1245http://dx.doi.org/10.1038/nrc2391http://dx.doi.org/10.3109/09553002.2010.537431http://dx.doi.org/10.1007/s00411-006-0088-6http://dx.doi.org/10.1088/1742-6596/41/1/047http://dx.doi.org/10.3109/0284186X.2011.582513http://dx.doi.org/10.1088/0031-9155/44/11/308http://dx.doi.org/10.1088/1367-2630/11/2/025001http://dx.doi.org/10.1126/science.327.5962.142http://dx.doi.org/10.1088/0031-9155/54/16/001http://dx.doi.org/10.1186/1748-717X-5-61http://dx.doi.org/10.1088/0031-9155/53/9/003http://dx.doi.org/10.1088/0031-9155/37/1/016http://dx.doi.org/10.1016/S0360-3016(00)00524-1http://dx.doi.org/10.1016/S0360-3016(00)00524-1http://dx.doi.org/10.1016/j.radonc.2004.02.007http://dx.doi.org/10.1088/0031-9155/29/5/008http://dx.doi.org/10.1016/j.radonc.2010.04.016http://dx.doi.org/10.1016/S0167-8140(02)00367-5http://dx.doi.org/10.1378/chest.126.4.1198http://dx.doi.org/10.1016/j.ijrobp.2005.04.022http://dx.doi.org/10.1016/j.ijrobp.2007.05.018http://dx.doi.org/10.1269/jrr.10016http://dx.doi.org/10.1016/j.ijrobp.2004.01.032http://dx.doi.org/10.1016/j.ijrobp.2004.01.032http://dx.doi.org/10.1158/1078-0432.CCR-04-1350http://dx.doi.org/10.3109/0284186X.2011.590148http://dx.doi.org/10.1097/JTO.0b013e3181560a68http://dx.doi.org/10.1097/IAE.0b013e3181dee621http://dx.doi.org/10.1097/IAE.0b013e3181ad2692http://dx.doi.org/10.1016/j.ophtha.2006.05.036http://dx.doi.org/10.1016/j.ophtha.2006.05.036http://dx.doi.org/10.1118/1.3291648http://dx.doi.org/10.2147/OPTHhttp://dx.doi.org/10.3109/08820538.2011.570846
  • 7/29/2019 Ions for Non Malignancies

    12/12

    Committee for Practice Guidelines, European Heart Rhythm Association,Heart Rhythm Society, ACC/AHA/ESC 2006 guidelines for the man-agement of patients with atrial fibrillation: full text: A report of the Amer-ican College of Cardiology/American Heart Association Task Force onpractice guidelines and the European Society of Cardiology Committeefor Practice Guidelines (Writing Committee to Revise the 2001 guide-lines for the management of patients with atrial fibrillation) developed incollaboration with the European Heart Rhythm Association and the HeartRhythm Society, Europace 8, 651745 (2006).

    85T. Terasawa, E. M. Balk, M. Chung, A. C. Garlitski, A. A. Alsheikh-Ali,J. Lau, and S. Ip, Systematic review: Comparative effectiveness of ra-diofrequency catheter ablation for atrial fibrillation, Ann. Intern. Med.151,191202 (2009).

    86A. Cheema, C. R. Vasamreddy, D. Dalal, J. E. Marine, J. Dong, C. A.Henrikson, D. Spragg, A. Cheng, S. Nazarian, S. Sinha, H. Halperin, R.Berger, and H. Calkins, Long-term single procedure efficacy of catheterablation of atrial fibrillation, J. Interv. Card. Electrophysiol. 15,145155 (2006).

    87R. M. Sullivan and A. Mazur, Stereotactic robotic radiosurgery(CyberHeart): A cyber revolution in cardiac ablation?, Heart Rhythm. 7,811812 (2010).

    88A. Sharma, D. Wong, G. Weidlich, T. Fogarty, A. Jack, T. Sumanaweera,and P. Maguire, Noninvasive stereotactic radiosurgery (CyberHeart) forcreation of ablation lesions in the atrium, Heart Rhythm. 7, 802810(2010).

    89M. Amino, K. Yoshioka, T. Tanabe, E. Tanaka, H. Mori, Y. Furusawa,W. Zareba, M. Yamazaki, H. Nakagawa, H. Honjo, K. Yasui, K. Kamiya,and I. Kodama, Heavy ion radition up-regulates Cx43 and amelioratesarrhythmogenic substrates in hearts after myocardial infarction, Cardio-vasc. Res. 72, 412421 (2006).

    90M. Amino, K. Yoshioka, D. Fujibayashi, T. Hashida, Y. Furusawa, W.Zareba, Y. Ikari, E. Tanaka, H. Mori, S. Inokuchi, I. Kodama, and T.Tanabe, Year-long upregulation of connexin43 in rabbit hearts by heavyion irradiation, Am. J. Physiol. Heart. Circ. Physiol. 298, H1014H1021(2010).

    91T. Elsasser, W. Kraft-Weyrather, T. Friedrich, M. Durante, G. Iancu, M.Kramer, G. Kragl, S. Brons, M. Winter, K. J. Weber, and M. Scholz,Quantification of the relative biological effectiveness for ion beamradiotherapy: Direct experimental comparison of proton and carbon ionbeams and a novel approach for treatment planning, Int. J. Radiat.Oncol., Biol., Phys. 78, 11771183 (2010).

    92L. Paulis and T. Unger, Novel therapeutic targets for hypertension,Nat. Rev. Cardiol. 7, 431441 (2010).

    93Symplicity HTN-2 Investigators, Renal sympathetic denervation inpatients with treatment-resistant hypertension (The Symplicity HTN-2Trial): A randomised controlled trial, Lancet 376, 19031909 (2010).

    94Symplicity HTN-1 Investigators, Catheter-based renal sympathetic de-nervation for resistant hypertension: durability of blood pressure reduc-tion out to 24 months, Hypertension 57, 911917 (2011).

    95E. J. Dropcho, Neurotoxicity of radiation therapy, Neurol. Clin. 28,217234 (2010).

    96S. Fridberg and B. I. Ruden, Hypofractionation in radiotherapy. Aninvestigation of injured Swedish women, treated for cancer of the breast,Acta Oncol. 48, 822831 (2009).

    97I. Suramo, M. Paivansalo, and V. Myllyla, Cranio-caudal movements ofthe liver, pancreas and kidneys in respiration, Acta Radiol. Diagn. 25,129131 (1984).

    98E. Rietzel and C. Bert, Respiratory motion management in particletherapy, Med. Phys. 37, 449460 (2010).

    99C. Bert, A. Gemmel, N. Saito, N. Chaudhri, D. Schardt, M. Durante, G.Kraft, and E. Rietzel, Dosimetric precision of an ion beam tracking sys-tem, Radiat Oncol. 5, 61 (2010).

    100T. Nomiya, H. Tsuji, N. Hirasawa, H. Kato, T. Kamada, J. Mizoe, H.Kishi, K. Kamura, H. Wada, K. Nemoto, and H. Tsujii, Carbon ion radi-ation therapy for primary renal cell carcinoma: initial clinical experi-ence, Int. J. Radiat. Oncol., Biol., Phys. 72, 828833 (2008).

    101Y. Aoka, T. Kamada, M. Kawana, Y. Yamada, T. Nishikawa, H. Kasa-nuki, and H. Tsujii, Primary cardiac angiosarcoma treated with carbon-ion radiotherapy, Lancet Oncol. 5, 636638 (2004).

    102S. M. Zenklusen, E. Pedroni, and D. A. Meer, Study on repainting strat-egies for treating moderately moving targets with proton pencil beamscanning at the new Gantry 2 at PSI, Phys. Med. Biol. 55, 51035121(2010).

    103C. Bert and E. Rietzel, 4D treatment planning for scanned ion beams,Radiat. Oncol. 2, 24 (2007).

    104R. Luchtenborg, N. Saito, M. Durante, and C. Bert, Experimental verifi-cation of a real-time compensation functionality for dose changes due totarget motion in scanned particle therapy, Med Phys. 38, 54485458(2011).

    105Y. Y. Vinogradskiy, P. Balter, D. S. Followill, P. E. Alvarez, R. A.White, and G. Starkschall, Verification of four-dimensional photon dosecalculations, Med. Phys. 36, 34383447 (2009).

    106D. S. Followill, D. R. Evans, C. Cherry, A. Molineu, G. Fisher, W. F.Hanson, and G. S. Ibbott, Design, development, and implementation ofthe radiological physics centers pelvis and thorax anthropomorphic qual-ity assurance phantoms, Med. Phys. 34, 20702076 (2007).

    107J. Biederer and M. Heller, Artificial thorax for MR imaging studies inporcine heart-lung preparations, Radiology 226, 250255 (2003).

    108K. Parodi, N. Saito, N. Chaudhri, C. Richter, M. Durante, W. Enghardt,E. Rietzel, and C. Bert, 4D in-beam positron emission tomography forverification of motion-compensated ion beam therapy, Med. Phys. 36,42304243 (2009).

    1727 Bert, Engenhart-Cabillic, and Durante: Particle therapy for noncancer d iseases 1727

    Medical Physics, Vol. 39, No. 4, April 2012

    http://dx.doi.org/10.1093/europace/eul097http://dx.doi.org/10.1007/s10840-006-9005-9http://dx.doi.org/10.1016/j.hrthm.2010.02.033http://dx.doi.org/10.1016/j.hrthm.2010.02.010http://dx.doi.org/10.1016/j.cardiores.2006.09.010http://dx.doi.org/10.1016/j.cardiores.2006.09.010http://dx.doi.org/10.1152/ajpheart.00160.2009http://dx.doi.org/10.1016/j.ijrobp.2010.05.014http://dx.doi.org/10.1016/j.ijrobp.2010.05.014http://dx.doi.org/10.1038/nrcardio.2010.85http://dx.doi.org/10.1016/S0140-6736(10)62039-9http://dx.doi.org/10.1161/HYPERTENSIONAHA.110.163014http://dx.doi.org/10.1016/j.ncl.2009.09.008http://dx.doi.org/10.1080/02841860902824917http://dx.doi.org/10.1118/1.3250856http://dx.doi.org/10.1186/1748-717X-5-61http://dx.doi.org/10.1016/j.ijrobp.2008.01.043http://dx.doi.org/10.1016/S1470-2045(04)01600-6http://dx.doi.org/10.1088/0031-9155/55/17/014http://dx.doi.org/10.1186/1748-717X-2-24http://dx.doi.org/10.1118/1.3633891http://dx.doi.org/10.1118/1.3157233http://dx.doi.org/10.1118/1.2737158http://dx.doi.org/10.1148/radiol.2261011275http://dx.doi.org/10.1118/1.3196236http://dx.doi.org/10.1118/1.3196236http://dx.doi.org/10.1148/radiol.2261011275http://dx.doi.org/10.1118/1.2737158http://dx.doi.org/10.1118/1.3157233http://dx.doi.org/10.1118/1.3633891http://dx.doi.org/10.1186/1748-717X-2-24http://dx.doi.org/10.1088/0031-9155/55/17/014http://dx.doi.org/10.1016/S1470-2045(04)01600-6http://dx.doi.org/10.1016/j.ijrobp.2008.01.043http://dx.doi.org/10.1186/1748-717X-5-61http://dx.doi.org/10.1118/1.3250856http://dx.doi.org/10.1080/02841860902824917http://dx.doi.org/10.1016/j.ncl.2009.09.008http://dx.doi.org/10.1161/HYPERTENSIONAHA.110.163014http://dx.doi.org/10.1016/S0140-6736(10)62039-9http://dx.doi.org/10.1038/nrcardio.2010.85http://dx.doi.org/10.1016/j.ijrobp.2010.05.014http://dx.doi.org/10.1016/j.ijrobp.2010.05.014http://dx.doi.org/10.1152/ajpheart.00160.2009http://dx.doi.org/10.1016/j.cardiores.2006.09.010http://dx.doi.org/10.1016/j.cardiores.2006.09.010http://dx.doi.org/10.1016/j.hrthm.2010.02.010http://dx.doi.org/10.1016/j.hrthm.2010.02.033http://dx.doi.org/10.1007/s10840-006-9005-9http://dx.doi.org/10.1093/europace/eul097