Está en la página 1de 16

Anniversary Paper: Past and current issues, and trends in brachytherapy physics

Bruce R. Thomadsena
Departments of Medical Physics, Human Oncology, Biomedical Engineering and Engineering Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705

Jeffrey F. Williamson
Department of Radiation Oncology, Virginia Commonwealth University, P.O. Box 980058, Richmond, Virginia 23298-0058

Mark J. Rivard
Department of Radiation Oncology, Tufts Medical Center, Tufts University School of Medicine, 750 Washington Street, #246, Boston, Massachusetts 02111

Ali S. Meigooni
Department of Radiation Medicine, University of Kentucky Medical Center, 800 Rose Street, Lexington, Kentucky 40536

Received 18 April 2008; revised 18 August 2008; accepted for publication 19 August 2008; published 24 September 2008 Brachytherapy began at the turn of the 20th century, contemporary with external-beam radiotherapy. Physicists and physicians together developed the eld. There has not been a period since the beginning that has not witnessed innovations and progress in brachytherapy. At the time of this article, the pace of change in the eld has never been more rapid, particularly in image-guided brachytherapy and the development of unconventional sources and treatment techniques. 2008 American Association of Physicists in Medicine. DOI: 10.1118/1.2981826 Key words: brachytherapy, dosimetry, history, trends I. MAJOR TRENDS AND CHALLENGES IN BRACHYTHERAPY Since its inception in the early 20th century, brachytherapy and its evolution have had a close association with medical physics. The 50 year history of the American Association of Physicists in Medicine AAPM has coincided not only with the emergence of medical physics as a mature profession, but with truly revolutionary innovations in radiological physics, including nuclear reactors, new particle accelerators, threedimensional 3D imaging, and computer-assisted treatment planning. Along with conceptual advances in radiation transport and modulation of clinical response, these developments have dramatically altered the practice of brachytherapy. Through fortunate intersections of technological opportunity and clinical need, brachytherapy has continued to serve an important, if rapidly changing, role in cancer management despite challenges to its survival from surgery and other treatment modalities. A recently published review and a 50th anniversary article have thoroughly covered the history of brachytherapy technology1 and dosimetry,1,2 an area in which the AAPM has had a major impact as an organization. Rather than duplicate or summarize these excellent reviews, we propose to focus this article on current trends and challenges in brachytherapy, particularly those that fall within the scope of medical physics practice or research activities. Probably the highest impact technological advances in the last half century were the introduction of articial radionuclides and afterloading into clinical brachytherapy.1 These advances signicantly lowered cost, reduced personnel expo4708 Med. Phys. 35 10, October 2008

sure, and increased technical exibility. They also set the stage for the renaissance of low dose-rate LDR temporary brachytherapy techniques beginning in the early 1960s in spite of the rapid penetration and development of megavoltage beam therapy, which made curative external-beam therapy feasible. Other important advances, reviewed elsewhere,2,3 were computer-assisted treatment planning/ dose evaluation and advances in dosimetry. Together, these advances set the stage for dramatic shifts in both clinical indications and technical practice that have occurred in the last 15 years.
I.A. New and evolving clinical applications

One remarkable trend has been the nearly exponential growth of transrectal ultrasound TRUS-guided brachytherapy for treatment of low and intermediate risk prostate cancer using low-activity 103Pd or 125I. Perhaps due to the attraction of a 1 day procedure along with a favorable prole of normal tissue complications, the number of procedures has grown from less than 5000 in 1995 to between 40 000 and 60 000 in 2002.4 This is approximately 30% to 40% of all eligible patients diagnosed annually in the United States, challenging radical prostatectomy as the standard of treatment. A second major growth area is breast conservation therapy, in which lumpectomy is followed by fractionated high dose-rate HDR brachytherapy 34 Gy in ten fractions administered over 5 days using either interstitial brachytherapy5,6 or a balloon applicator7 in place of 6 weeks of external-beam therapy. A third clinical indication for a
2008 Am. Assoc. Phys. Med. 4708

0094-2405/2008/3510/4708/16/$23.00

4709

Thomadsen et al.: Brachy Anniversary Paper

4709

new brachytherapy application is intravascular brachytherapy IVBT, which was introduced in the late 1990s with much excitement in the radiation oncology community. Randomized clinical trials soon showed that IVBT dramatically lowered the incidence of restenosis following percutaneous coronary angioplasty relative to unirradiated controls.8 As many as 40 000 intravascular brachytherapy procedures were performed annually as its utilization peaked in 2002. Perhaps due to unequivocally positive results only for in-stent restenosis, late stent thrombosis, and recurrent restenosis at the edge of the treated eld,9 IVBT was abandoned as rapidly as it was developed in favor of rapamycin- and paclitaxeleluting stents.10,11 This remarkable phenomenon illustrates how vulnerable specialized surgical procedures practiced by nonsurgeons, e.g., brachytherapists, are to competing surgical technologies. Growth in the areas of permanent prostate brachytherapy and HDR 192Ir accelerated partial breast irradiation has been accompanied by a reduction in the use of temporary LDR brachytherapy at other sites. This reduction is due, in part, to migration of traditional LDR brachytherapy procedures, e.g., intracavitary brachytherapy for gynecological malignancies, to fractionated HDR. At least in the United State, use of denitive brachytherapy for sites such as early stage head and neck has failed to compete effectively with other therapeutic options, including intensity-modulated radiation therapy IMRT and surgery.
I.B. Physics and technical innovations in brachytherapy

planning used to correct source position.16,17 On the horizon, positron emission tomography and other biological imaging modalities should work its way into brachytherapy treatment planning as it has in external-beam radiotherapy.18 I.B.2. Improved dosimetry and treatment planning The rapidly growing utilization of low-energy interstitial sources has motivated intensive investigation of dosimetry techniques for more accurately estimating clinical dose distributions, a development that has greatly beneted from AAPMs leadership and consensus building.2 Parallel developments in computational and experimental dosimetry3 have reduced physical dose specication uncertainty to the 3% to 5% level k = 1 for 125I brachytherapy.19 Another emerging development is the application of Monte Carlo-based dose calculations to treatment planning,20,21 which will eliminate major uncertainties in clinical dose specication due to tissue heterogeneities, interseed attenuation, and applicator shielding effects.
I.C. Scientic and clinical challenges facing brachytherapy

I.B.1. Integration of 3D medical imaging into brachytherapy Throughout most of its long life, brachytherapy has evolved as a surgical art, in which seed or applicator positioning was guided by palpation or visualization of the target tissue. Treatment planning consisted of calculating dose relative to the source positions derived from orthogonal radiographs, not the underlying anatomy. While a number of pioneering publications investigated the registration of dose distributions to 3D medical images throughout the 1980s and 1990s, only with the rise of prostate seed brachytherapy did image-guided source placement or image-based planning achieve widespread use in brachytherapy. TRUS-guided seed placement and post-implant x-ray and computer-aided tomography CT imaging, used to evaluate the nal dose distribution, are now standards of practice.12 CT-based catheter guidance and dose evaluation is commonplace for partial breast brachytherapy,13 and magnetic resonance imaging is emerging as the planning image modality of choice for gynecological tumors.14 These exciting developments offer many potential advantages, including a planning process conceptually similar to that of teletherapy, less dependence on surgical skill, and most importantly, enhanced targeting accuracy, target conformality, and normal tissue avoidance. Associated developments include intraoperative planning planning and delivery integrated into a single procedure15 and intraoperative adaptive planning intraoperative dose reMedical Physics, Vol. 35, No. 10, October 2008

While brachytherapy usually is an invasive procedure limited to surgically accessible tumor sites, it is able to safely deliver much higher biological equivalent doses than IMRT.22 HDR brachytherapy is able to deliver very large fraction sizes safely while, at the other end of the spectrum, permanent seed brachytherapy the ultimate form of hyperfractionation supports delivery of very high physical doses due to repair of normal tissue sublethal damage. Credit is often given to brachytherapys superior targeting accuracy and conformality direct insertion of sources into target tissue versus external ducial alignment and minimum impact of tissue motion, which is widely assumed to make planning target volume PTV expansions unnecessary. For brachytherapy to retain these competitive advantages, the following scientic issues require urgent attention. I.C.1. Improved understanding and exploitation of geometric and radiobiological uncertainties Geometric uncertainty, and its role in determining PTV margins, has been extensively studied in external-beam radiation therapy in contrast to brachytherapy,23 where relatively little data is available.22 Image-guided radiotherapy IGRT techniques have markedly improved external-beam precision.24 Brachytherapy-like hypofractionated regimens have been successfully delivered to patients with lung25 and prostate cancers26 using external beam IGRT. To use brachytherapy safely for highly conformal treatment of well-dened target volumes, a better understanding of the interplay between organ delineation errors, seed and applicator positioning uncertainties, and intrafractional tissue motion are needed. For permanent brachytherapy, a better understanding of normal and target-tissue response to the ultralow dose rates, along with the modulating effects of geometric uncer-

4710

Thomadsen et al.: Brachy Anniversary Paper

4710

tainties, are required to be able to extend permanent seed brachytherapy to new treatment sites without mounting doseseeking phase I and II clinical trials. I.C.2. Improved management of tissue deformation Since brachytherapy is a surgical intervention, it deforms and displaces tissues in a highly localized and nonlinear fashion. These problems hinder accurate registration of pretreatment biological images onto intraoperatively available images or preclude accurate summation of biological doses from individual brachytherapy fractions. Deformable image registration is a promising solution to this problem and has been demonstrated in both prostate27 and gynecological28 brachytherapy applications. However, many challenging problems remain to be solved, including general and patientspecic validation of such registrations.29 I.C.3. More realistic dose-computation techniques Despite substantial progress in single-source brachytherapy dosimetry, current table-based dose computation algorithms model patients as uniform water spheres of water, neglecting tissue density and composition heterogeneities and applicator- and source-shielding effects. This approximation results in large differences between the dose distribution actually delivered to the patient and that calculated by the treatment planning system, particularly for low-energy sources. This topic is discussed below. II. BRACHYTHERAPY DOSIMETRY
II.A. Brachytherapy treatment planning calculation algorithms

Although discussed briey here, the reader is directed to the references cited for discussions of each of the parameters. The 2D formalism tends to reproduce actual dose distributions more realistically than the 1D formalism, especially for points located near the source long-axis. As of this writing, the 2D formalism is used almost exclusively for HDR brachytherapy treatment planning, where a single source is used and conned to a route dened by a catheter or needle, or temporary LDR interstitial implants. The 1D formalism is used most frequently for permanent LDR brachytherapy treatment planning, where the active source length is less than about 0.5 cm and the orientation of individual sources cannot be determined accurately. Practical considerations often prohibit the use of the 2D formalism in permanent implant cases. II.A.1. Source Strength SK Currently the recommendation for expressing source strength is in terms of air-kerma strength, equal to the product of the air-kerma rate for a small mass of air in vacuum and the distance squared, in units of Gy h1 m2 = cGy h1 cm2.39 For convenience, the units are abbreviated as U. Considerable improvements have been made in reducing the uncertainties of source calibrations over recent years, and continue through the time of writing see, for example, Rasmussen et al.40,41. While most of the efforts have been directed toward improving the determination of SK, calorimetric approaches have also been investigated that would yield the power emitted by a source or the dose rate to a point.42,43 II.A.2. Dose rate constant The dose rate constant is dened as the dose rate per unit air-kerma strength at a reference point along the transverse axis, ro = 1 cm and = 90 or / 2 radians of the source: r , /2/S , =D o K 2

One of the greatest contributions of the AAPM to radiotherapy physics has been the development of brachytherapy source dosimetry. Based on work by Meisberger and Dale,3032 the current established method for brachytherapy treatment planning uses the formalism introduced by the Interstitial Collaborative Working Group33 and rened by AAPM Task Group No. 43 report.34 In 2004, this protocol was updated to eliminate shortcomings due to the lack of information when the original protocol was published.19 The TG-43 formalism has been applied to dose calculations around the intravascular brachytherapy sources,35,36 elongated brachytherapy sources,37 and 137Cs sources.38 In the TG-43 protocol, the dose rate distribution around a sealed brachytherapy source can be determined in two dimensions 2D using the following formalism: r , = S K G r , g r F r , D L L G r o, / 2 1a

where ro customarily falls at 1 cm for photon sources, or 2 mm for beta emitters. Predicting the value for the dose rate constant from spectroscopic measurements has proven reliable for many sources and shows some promise for replacing direct measurement.44 II.A.3. Radial dose function, gr and the geometry function Gr , The radial dose function gr represents the attenuation of the radiation in tissue, dened as gr = r, /2 Gr , /2 D o , r , /2 Gr, /2 D
o

or without knowledge of the source orientation, a onedimensional 1D version r = S K g r r , D p an r2 1b

where r is the distance from the source center to the point of interest, and is the angle with respect to the source axis.
Medical Physics, Vol. 35, No. 10, October 2008

where Gr , is the geometry function which accounts for the effect of the distribution of radioactive material inside the source on the dose distribution at a given point. The geometry function is dened as34

4711

Thomadsen et al.: Brachy Anniversary Paper

4711

Gr, =

1/r2 ,
1 1

Point source approximation

tan x + L/2/y tan x L/2/y Linear source approximation 0 Ly 1 Linear source approximation = 0 and x L/2 2 x L/22

where x and y are the coordinates of the point of interest relative to the longitudinal and transverse axes of the source, and L is the active length of the source. As seen in Eq. 3, the value of the radial dose function is equal to 1 at the reference point along that sources transverse bisector, ro , / 2. An update to the TG 43 report claried the length of the source when the construction consists of spheres or pellets instead of a linear carrier as an effective length Leff equal to the number of pellets times the centerto-center distance between the sources.19 II.A.4. 2D Anisotropy Function Fr , The 2D anisotropy function Fr , represents the variation of the dose distribution around a brachytherapy source due to the distribution of radioactivity within the source, self-absorption, and oblique ltration of the radiation in the capsule material. The TG-43 report denes the anisotropy function as Fr, = r, Gr, /2 D . r, /2 Gr, D 5

II.A.5. 1D Anisotropy Function anr The 1D anisotropy function, or anisotropy factor anr for a given radial distance comes from integrating the dose rates over the polar angle assuming a cylindrical source at a given radial distance:

anr =

Dr, sind 0 . r, 2D
0

II.B. Clinical dose calculations

Clinical use of computerized systems has permitted dosimetry data obtained by researchers to overlay source dose distributions, via the superposition principle, onto patientspecic geometries. Use of this principle becomes inappropriate when the interactions between sources become signicant, when the geometry used by the dosimetry investigator does not approximate well that of the patient, and when differences between the patients tissue and the water medium used with TG-43 tables become pronounced. Dose distributions obtained by researchers for a single source at specic points are tted to mathematical functions for entry into a radiotherapy treatment planning system and subsequent derivation of clinical dose distributions. This data
Medical Physics, Vol. 35, No. 10, October 2008

tting is most typically performed for the radial dose function where it is impossible for the brachytherapy dosimetry researcher to determine the dose distribution at all possible clinical locations. There is a wide assortment of possible techniques for clinical dose calculations. Some planning systems utilize a polynomial t while others prompt the clinical physicist to enter data with a given bin size or spatial resolution, sometimes requiring specication of dose at the source origin or within the source capsule! Clearly, differences in dose distributions can arise when there are differences in the choice of tting functions between the dosimetry investigator and the clinical treatment planning system. To minimize the likelihood of this occurrence, the AAPM has in place a mechanism for issuing consensus datasets for brachytherapy source dose distributions. The AAPM Low- and High-Energy Brachytherapy Source Dosimetry Working Groups under the Brachytherapy Subcommittee perform this function and typically publish recommended consensus datasets and dosimetry formalisms in the journal Medical Physics. These datasets include the following source-model specic dosimetry parameters: effective Leff or active source length L; dose rate constant ; radial dose function for the point- g Pr, or line-source gLr, approximation; the 2D anisotropy function Fr , ; and 1D anisotropy function anr. The range of data specied by the dosimetry investigator is often inadequate to cover all clinical possibilities. Consequently, and to promote uniform brachytherapy source delivery and patient treatment, the AAPM has further recommended interpolation and extrapolation techniques to facilitate brachytherapy dose calculation beyond the range published by the dosimetry investigator.19 Not all vendors of brachytherapy treatment planning systems have incorporated the AAPM recommendations. The largest dose differences may arise at locations close to the source, near the source long-axis, or in between researcher-specied points.19 In addition to concerns with dosimetry parameters, brachytherapy physicists should be concerned with the grid size used for dose calculationboth upon initial source characterization and subsequent treatment planning. Some planning systems permit the physicist to set grid size, and detrimental effects can result if it is set too large. For example, Fig. 1 shows the impact of setting 0.1, 1, 2, and 5 mm grid size during treatment planning for the same 125I source. The planning systems did not change the dose calculated to the 15 circular points or to dose volume histograms when changing the grid size, but visualization of isodose rate distribu-

4712

Thomadsen et al.: Brachy Anniversary Paper

4712

converted to absorbed dose using the following formalism, described by Meigooni et al.,45 and Williamson and Rivard,3 r, = D Rnetr, , SKErdT FlinT 7

FIG. 1. Impact of dose grid size on visualization isodose rate distributions. The isodose lines decrease from 10, 5, 2, 1, 0.5, and 0.2 cGy / h for a 0.5 U model 6711 I source. The radioactive component is indicated by the centrally positioned rectangle within the source capsule. From upper left to lower right, the grid size is 5, 2, and 1 mm, and 0.1 mm on a side. Scale ticks indicate 10 mm.

tions became perturbed as grid size increased. The physicist must know the limitations of the planning system before setting out to calculate clinical dose distributions and deciding on the optimal grid size to balance dose calculation time with dose calculation accuracy.
II.C. Measurement of dosimetric parameters

Due to high dose gradients and small dose rates around brachytherapy sources, measurement of dose distribution is labor intensive, particularly for low-energy photon-emitting sources. A detector with a wide dynamic range, at energy response, small geometric dimensions, and high sensitivity is a suitable device for these measurements. The low signal-tonoise ratio in this eld limits application of ionization chambers in these measurements. Currently, LiF thermoluminescent dosimeters TLDs are the most commonly used detectors in this application. Silicon diodes, radiochromic lm, and diamond detectors also have been used. In this section, the main emphasis is on measurement of the dose distribution around brachytherapy sources with LiF TLDs because most measurements of the dosimetric parameters for brachytherapy sources have been made using these dosimeters. LiF TLDs are available in various forms and shapes; however, chips of 3.1 3.1 0.8 mm3 and 1.0 1.0 1.0 mm3 are the two most commonly used. For dose measurements with these detectors, slabs of water equivalent phantom material can be machined accurately to accommodate the source and the TLD. Care must be taken to avoid dosimeters eclipsing more distant dosimeters during the measurements. Responses of the irradiated TLD in a given measurement are
Medical Physics, Vol. 35, No. 10, October 2008

r , is the dose rate at a point r , , R r , is the where D net net response of the TLDs placed at r , as corrected for background and the physical differences among TLD chips using predetermined chip calibrations.45 T is the irradiation duration, SK is the measured source air-kerma strength at the time of measurement, and is the calibration factor for the TLD response e.g., nC/cGy usually using a calibrated Co60, 4 MV, or 6 MV photon beam. Occasionally supercial x-ray beams are used for TLD calibration. Er is a correction factor for the energy dependence of the TLD between the calibration beam and the brachytherapy source; for instance, a value of 1.4 for both 125I and 103Pd source dosimetry if the TLDs are calibrated with a megavoltage beam.3,46 dT is a correction factor used to account for the decay of the source during the experiment, and Flin accounts for TLD nonlinearity. For measurement of the dose rate constant, TLDs are placed 1 cm from the axis of the source on the perpendicular bisector at a number of angles around the source. TLD measurements of radial dose function are normally performed at distances ranging from 0.5 to 10 cm at 0.5 and 1 cm increments. The measured value at each distance is obtained from the average of several TLD chips to achieve an uncertainty one standard deviation of about 4%. Determination of the anisotropy function requires measurements at many angles and distances with the measurements taken several times to allow averaging to achieve the 4% uncertainty.19 Because the use of TLDs, or any point dosimeter, requires so many individual measurements, planar detectors such as radiochromic lm become attractive. The calibration, use, and reading of the lm all pose potential problems and require consideration of details.4750 While gel dosimetry looks appealing because it conceptually could provide an entire set of three-dimensional data with a single exposure, characteristics such as the high dose required for adequate signals and a marked gradient effect pose challenges for widespread use.5155
II.D. Monte Carlo simulations for estimating brachytherapy dosimetry

In the rst half of the 20th century up until the early years of the AAPM in the 1960s, clinical brachytherapy dose distributions were generally determined using lookup tables i.e., along-away tables based on ionization measurements and Sievert integrals.5658 Based on modeling brachytherapy source dose distributions in solid-state media using radiation transport, one of the earliest publications using Monte Carlo methods to simulate a geometrically realistic cylindrically symmetric brachytherapy source was by Krishnaswamy in 1971.59 This work and subsequent efforts set the stage for modern brachytherapy dosimetry simulations using Monte Carlo methods. Given Moores Law for exponential increase

4713

Thomadsen et al.: Brachy Anniversary Paper MCNPX, Williamsons PTRAN GEANT4,


86 87

4713

in computer processor speed,60 the 5 107 histories for the 252 Cf applicator tube source performed in 1997 by Rivard et al. closely followed the expected increase in comparison to the 5 103 histories performed in 1970 by Krishnaswamy over a timeframe of about three decades.59,61 Based on the Monte Carlo simulations by Krishnaswamy, over a decade passed before other investigators simulated brachytherapy sources to the same level of detail as Krishnaswamy. For example, isotropic point-source build-up factor-based approaches to brachytherapy dose distribution derivation were performed by Berger,62 Webb and Fox,63 and in the famous paper by Meisberger et al.30 More sophisticated approaches using 3D modeling were pursued later, and were largely spearheaded by Williamson.6470 Various Monte Carlo codes for radiation i.e., 0.03 to 1 MeV photon transport had been used since the 1950s, but medical physicists transcended the gap between basic physics and the needs of the clinic, often by developing homemade radiation transport codes.7173 Once brachytherapy source dose distributions were calculated, these results were readily integrated into then-modern approaches for treatment planning.58,74 In fact, the rst president of the AAPM, Gail Adams, was involved,75,76 along with other groups Nelson,77 Hope,78 Shalek79,80 in developing computer-assisted dose calculations based upon precomputed Sievert integral single-source dose distributions and the superposition principle. A detailed summary of this era was prepared by Stovall and Shalek.58 With increases in processor speed, accessibility of operating systems, and more widespread use across medical research, Monte Carlo methods for simulating brachytherapy dose distributions reached a critical threshold in the 1990s. A key event was the publication of the 1995 AAPM TG-43 report.34 This report signicantly advanced the standard brachytherapy dosimetry formalism, required separate brachytherapy dosimetry parameters for different source models of the same radionuclide, promoted consistent use of brachytherapy dosimetry parameters at separate institutions, and through its popularity, informed medical physicists far and wide of the possibilities of Monte Carlo methods for simulating brachytherapy source dose distributions. Monte Carlo methods for brachytherapy dosimetry were further popularized by Williamson using his PTRAN code, where different source types and aspects of brachytherapy were examined.81 Monte Carlo codes are products of humans, and are consequently subject to a variety of errors. For example, some studies published on Monte Carlo-derived brachytherapy dosimetry methods accidentally82 utilized photon cross-section libraries based on antiquated data.83 DeMarco and colleagues rst noticed this problem for the MCNP code.84 Subsequently, modern cross-section libraries85 have been adopted by groups using established Monte Carlo codes. The AAPM methodologically examined the role of Monte Carlo codes for simulating brachytherapy source dose distributions.19 Specically, Monte Carlo radiation transport codes were identied that have been well benchmarked for brachytherapy dosimetry. These included EGS, MCNP, PTRAN
Medical Physics, Vol. 35, No. 10, October 2008

and EGSnrc.88 Only EGS, MCNP, and code were mentioned in the TG-43U1 report as examples of well-benchmarked codes. The eld of Monte Carlo simulation of brachytherapy source dose distributions is mature; however, there is room for improvement towards better simulating the clinical environment. Since Krishnaswamy, it has been shown that improved dose simulation accuracy, determined through comparisons with measurements, are obtained through accurate modeling of the brachytherapy source and capsule. This approach has been extended to investigation of interseed interactions8991 and motion of internal components.91 In fact, the principal equipment used to produce calibration results, i.e., the wide-angle free-air chamber at the National Institute of Standards and Technology,92 has been simulated for direct comparison with experimental measurements.93 Further advances include simulations of material heterogeneities,87,94 scatter conditions,86,95 and localization in the patient through image-guidance.94 With the introduction of HDR brachytherapy sources having relatively lowenergy photon emissions,87,9698 distant prescription points are more likely to cause dosimetric changes if accounting for material heterogeneities in comparison to assuming a liquid water environment. One must exercise caution, however, since a feature unique to Monte Carlo methods in comparison to measurement techniques is the capability to simulate unreal circumstances, for example, using water with a mass density of 10 g / mm3. In addition, Monte Carlo results, unlike experimental measurements, cannot account for modeling errors, e.g., contamination of radionuclide spectrum, errors in implementing calibration standards, etc. Comparison of Monte Carlo and limited experimental results is needed to detect unanticipated discrepancies between the assumed and actual properties of the system under investigation. Efforts are underway to incorporate Monte Carlo methods into clinical brachytherapy treatment planning systems due to their proven utility to simulate radiation scatter conditions and account for material heterogeneities.20,99 Other approaches not using Monte Carlo methods, such as the collapsed cone employing superposition of primary and scatter dose, may also account for dosimetric effects not modeled by the AAPM TG-43 dosimetry formalism.100,101 Thus, it remains to be seen whether or not Monte Carlo methods for brachytherapy dosimetry will transition from a research tool to the backbone of the treatment planning algorithm. Investigation of a similar shift occurred a few years ago for external beam treatment planning.102 However, the competing standard treatment planning algorithms for external beam radiotherapy provide much better agreement with measurements than the TG-43 dosimetry formalism for measurements of brachytherapy dose under challenging scatter and material heterogeneity conditions.
II.E. Accounting for heterogeneities in dose calculations

It is widely recognized that current dose-specication and -calculation practices introduce signicant dose-

4714

Thomadsen et al.: Brachy Anniversary Paper

4714

calculation errors by failing to adequately account for applicator attenuation, tissue heterogeneities, and intersource shielding. Some applicators containing local shielding to spare bladder and rectal tissues can produce dose reductions as large as 50%103,104 that most treatment planning systems still continue to ignore. The perturbing effects of partial transmission shielding have been shown73,105,106 to vary rapidly with cross-sectional area and location of shielding materials, especially for sources with energies at or below that of 192Ir. Only for 137Cs shielded sources can one-dimensional pathlength algorithms available on some commercial planning systems, e.g., generalizations of the Sievert integral model,107 be used reliably.108 These studies also document that Monte Carlo simulations accurately predict such effects. Seed-to-seed attenuation effects have been shown to produce moderate but potentially signicant systematic dose overestimates 2% to 4% average errors, and up to 15% local errors for 103Pd and 125I volume implants.90,109 For 125I and 103Pd sources, tissue-composition heterogeneities are known to have a high impact on theoretical grounds, as the photoelectric effect dominates energy deposition. Dose rates in adipose and fat-like tissues can deviate from water-equivalent calculations by as much as 50%,110,111 whereas in bony tissues, or soft tissues with signicant mineral content, conventional calculations underestimate true dose by as much as a factor of 2.112 On the other hand, replacement of as little as 1% of soft tissue mass by calcied tissue has been shown to reduce prostate doses by 8%.20 Maughans combustion analysis measurements show that mineral ash content of excised tumors ranges from 0.9% to 3%, leading to dose estimation errors as large as 15% at 28 keV.113 For higher energy sources, there is evidence that air-tissue boundaries forming the breast and lung contours can perturb HDR breast implant doses by 5% to 10% at lower doses 60% of D90 for 192Ir while for lower energy 169 Yb, D90 is lower than predicted by conventional dose planning by 5% and as much as 25% at lower doses.87 No studies systematically assessing the impact of compositional in contrast to density heterogeneities on higher energy sources are available. For 192Ir, Anagnostopoulos found that bony structures perturbed doses by up to 15% at low doses while Ye found that a 5% variation in contrast-agent concentration in the MammoSite applicator reduced dose by as much 7%.114,115 1D pathlength and other heuristic algorithms have been reviewed elsewhere.108 A more general approach is the superposition/convolution algorithm.21,116 Tedgrens algorithm, which adapts the collapsed cone methodology from external beam dose calculation, is being considered for implementation in the Nucletron PLATO planning system. Preliminary results suggest that superposition is quite accurate but computationally intensive. Another and more general deterministic algorithm is discrete ordinates, which rigorously solves the Boltzmann transport equation on a systematically discretized phase-space grid.117119 These solutions approach the complexity, rigor, and computational efciency of Monte Carlo simulation. Daskalov et al.118,120 have shown that single-processor 2D and 3D discrete ordinates calculaMedical Physics, Vol. 35, No. 10, October 2008

tions are one to two orders of magnitude faster than general purpose Monte Carlo code calculations of equivalent dimensionality, while Giffords code required 20 min of computing time for 3D dose distributions from a shielded colpostat.119 Both Daskalov and Gifford required analytic or Monte Carlo computation of the rst collision source to avoid ray effects characteristic of discrete point sources. Several groups are investigating direct application of Monte Carlo simulation for dose computation on clinically realistic multiple-source implants. Early studies required several hours or even days of computing time.90,121,122 However, two groups have reported Monte Carlo codes especially designed to facilitate efcient and more accurate planning in brachytherapy with 20 s to 1 min computation times for clinical implants.20,88 Because Monte Carlo calculations can be parallelized using PC clusters with distributed memory in a cost effective manner, in contrast to more expensive shared-memory multiple processors needed for discrete ordinates, the authors predict that Monte Carlo simulation may be the dominant brachytherapy dose-calculation algorithm of the future. Lack of voxel-by-voxel knowledge of the photoelectriceffect and scattering cross sections versus photon energy limits implementing any realistic dose-calculation algorithm for low-energy seeds. Although average compositions of human tissues and organs have been estimated,123 no data on variations within individual organs or from patient to patient are available. Quantitative dual-energy CT imaging is one possibility that has been investigated for bone-mineral assay124 and, early in the history of CT, for estimation of human tissue composition.125 While preliminary data suggest that dual-energy CT cross-section imaging is feasible,126,127 the high quality images required by this demanding application are difcult to achieve at reasonable patient doses on commercial scanners with currently available data preprocessing and reconstruction algorithms.
II.F. Optimization

Treatment planning for brachytherapy consists mostly of determining the catheter, needle, applicator or source locations, and the strength or dwell time for the sources used. Systems such as the Manchester128 or Paris129 give good suggestions for needle placement, as does the work of Kwan et al.130 and of Zwicker et al.131 Some general principles help guide needle location selection for interstitial implants: 1. To achieve the most uniform dose distributions, the concentration of sources or dwell positions near the periphery needs to be higher than in the interior. For many congurations, this approximates 75% of the source material or dwell weight being on the periphery, as suggested by the Manchester System132135 The actual percentage that should reside on the periphery depends on the shape of the CTV and the source positions. Needles in corners of implants should be moved inward to avoid breaks in the prescription isodose surface between sources.136 Reducing the distance to each of the nearest needles to about two-thirds of what it would be

2.

4715

Thomadsen et al.: Brachy Anniversary Paper

4715

3.

for a regular geometric pattern usually sufces. While most useful for uniformly loaded needles or catheters, moving the catheters inward in implants that will be optimized reduces the potential for large high dose volumes. When using small sources such as iridium seeds as opposed to wire, the strength at the ends of needles should be roughly twice that of each of the rest of the sources in the needle. Such loading helps prevent the prescription dose distribution from dipping into the treatment volume between the catheters.

target, target dose homogeneity, and doses to organs at risk, possibly in nonlinear combinations. II.F.1. Deterministic This class of optimization approaches refers to methods that produce the same result each time they run on the same case. Several deterministic approaches have been used for brachytherapy optimization, but the most successful and practical for permanent implants is branch and bound.140,141 In this technique, the strength of sources at all possible positions is solved in an analytical manner, giving an idyllic solution but with fractional sources at each position. One by one, the process considers each position and changes the fraction to a source present or absent, and reevaluates the objective function. The pattern of the changes guarantees a relatively efcient search not having to consider every option, and guarantees nding the absolute optimum solution. If simply satisfactory solutions are needed, the process becomes very rapid. A subset of deterministic approaches falls under the classication of analytical techniques. Used mostly for HDR applications, an analytical approach sets up a set of simultaneous equations where each equation expresses the dose at a point as a function of all the source dwell times that form the variables. For implants with a few hundred dwell positions and a similar number of points dening the dose to a target surface, the problem becomes very time consuming, and polynomial models and approximations are common. Because it solves for a particular set of constraints the doses to a set of points, this approach is not truly optimization, but satisfaction. II.F.2. Stochastic Stemming originally from permanent implants, simulated annealing and genetic simulation are the most common stochastic approaches. The techniques start with any potential binary loading pattern, then make changes and evaluate whether the new pattern is better than the original. At rst, the changes are major, but as the process progresses, the changes become more rened. Occasional large random changes help prevent the solution from settling into local optima. While not necessarily nding the global minimum of the objective function in a nite computing time, the stochastic approaches are likely to nd satisfactory solutions. The methodology now applies to HDR brachytherapy as well as permanent implants.142 II.F.3. Heuristics Instead of trying to nd optimal solutions, heuristics use very fast methods to nd useful solutions, often using surrogates for the dose distribution to guide the search. Geometric optimization, used mostly for HDR applications, weights each dwell time inversely to the sum of the distances between a given dwell position and all other dwell positions. The adjoint/greedy heuristic uses the adjoint sensitivity function to determine the most efcacious positions for source strength to provide a uniform dose to a target and minimize

Differential loading of implants can create a more uniform dose distribution,137 while a more uniform loading produces a higher dose in the center of the implanted volume. Either pattern may be consistent with the desired goals of the treatment. Intracavitary brachytherapy presents less opportunity to control the dose distribution, but the design of the applicators has a great impact on the resultant dose distribution. Some general principles for source distribution for intracavitary applications include the following: 1. When the goal is to project the dose to points distant from the treatment appliance, the source locations should be far from the applicator-tissue interface. Just as in external-beam radiotherapy, a greater source-tosurface distance gives a greater fractional depth dose, so also with brachytherapy. If possible, the applicator should place sources distributed around the target such as using tandem and ovoids rather than a tandem and cylinders. Recognize that the dose distribution will not be uniform, but continually decreasing with distance from the sources.

2.

3.

Determining the source strengths at each location for an LDR implant, or the source dwell times for an HDR 192Ir applicator, forms one of the problems most often identied as optimization. The LDR problem may be a quantized problem, such as which of eight source strength options should be placed at each source location, or a binary selection, for example in prostate implants, whether a source occupies a given location or not. The HDR problem is almost continuous, where the dwell times could assume any value between 0 and 999 min in 0.1 s increments. For a comprehensive discussion on optimization, see Ezzell138 and also Ezzell and Luthmann.139 Many techniques for optimization have found their way into brachytherapy, but because of space limitations, only those most commonly encountered or offering some unique advantage will be discussed here. The classications carry considerable arbitrariness, and several approaches sometimes could be listed under more than one classication. The list below only considers the approaches applied in brachytherapy in commercial systems or in recurring research articles. Many of the techniques use objective functions which evaluate how closely the solution matches the desired distribution. The functions often consider the dose coverage of the
Medical Physics, Vol. 35, No. 10, October 2008

4716

Thomadsen et al.: Brachy Anniversary Paper

4716

the dose to sensitive structures.143 The heuristics speed comes from the absence of iteration. One of the current challenges for optimization is livetime reoptimization guidance intraoperatively.16 This process would update the dose distribution as sources are placed, usually in a permanent implant, and recalculate the positions of the remaining sources. Such a process requires extremely fast computations and high quality imaging. Limitations on the length for this article prevent consideration of evaluation procedures for the quality of brachytherapy plans. Thomadsen has presented detailed discussion of plan evaluation.144

founded on MR images.14 While the use of MRI for treatment planning does present challenges, the future of cervical brachytherapy lies in this direction.
III.B. Prostate implants

III. BRACHYTHERAPY PROCEDURES: APPLICATIONS, APPROACHES, AND APPLICATORS


III.A. Cervix brachytherapy

Reports of brachytherapy for cancer of the uterine cervix trace back to at least 1903, and it soon became the preferred treatment modality.145 The early techniques varied greatly, mostly specifying treatment in terms of mg- h. The practice at the Holt Radium Institute in Manchester developed a system for treating patients designed to deliver the same dose to the control point, called Point A, a xed distance 2 cm superior to the vaginal fornices and 2 cm lateral to the uterine canal. This system found great acceptance throughout the world and formed the basis for much of cervical intracavitary brachytherapy to this day. At M.D. Anderson, Fletcher developed an approach calling for loading the radioactive material in the applicators based on the size of the applicator accommodated and on the stage of the disease.146 For the given combination of stage and applicator, the treatment would be specied as a particular number of hours, although limited to a specied total number of mg- h or dose to the bladder, rectum, or vaginal surface. In Paris, a different approach evolved particularly for preoperative brachytherapy, matching the 60 Gy isodose surface the reference isodose to the dimensions of the cervix and lower uterus.129 The introduction of HDR brachytherapy for cervical brachytherapy improved the accuracy of radiation delivery through optimization of the treatment plan and reduction in applicator movement during the treatment time. The HDR technique also reduced the unnecessary radiation exposure to medical staff, reduced the treatment duration time, and eliminated the mandatory hospitalization. All these methods used radiographs to determine the geometry of the applicator and visual inspection and palpation to establish the diameter of the cervix. The tumor played little or no role in dening the treatment. The advent of CT enabled better dosimetry for the bladder and rectum, but failed to visualize the tumor as distinct from the rest of the cervix. MRI, on the other hand, distinguishes between a tumor and normal cervix. The Group Europen de Curiethrapie of the European Society for Therapeutic Radiology and Oncology has developed a protocol for threedimensional, target-based intracavitary brachytherapy
Medical Physics, Vol. 35, No. 10, October 2008

Brachytherapy for treatment of prostate cancer has been used for about 100 years.147 Since those rst investigations with 226Ra needles, there have been many variations on implant technique and radionuclides implanted. Signicant advances since then include transperineal implantation using transrectal guided ultrasound, use of hormones to decrease gland size, widespread use of prostate-specic antigen testing to assist staging and treatment modality choice, use of radionuclides having lower photon energies than 226Ra, and the treatment planning advances described above. Since an excellent review of the current status of prostate brachytherapy is given in Chapters 28 through 33 of the 2005 joint AAPM/ABS Summer School text,148153 this section will focus on steps medical physicists are taking towards advancing this treatment technique. In this decade, there have been substantial advances to brachytherapy source dosimetry. These advances have come about in part due to increasing choices of source types and congurations. It is understood that some sources have dynamic internal components which can vary prostate dose depending on patient positioning/orientation.91 There are now seeds having nonmetallic plastic capsules,154,155 unencapsulated 103Pd wires,156158 seeds which may be connected together like LEGOs,159 and seeds not having cylindrically symmetric dose distributions that provide directional emissions for preferentially protecting critical structures.160 Further, seeds may now be purchased already contained in strands or needles, complicating direct calibration.161 This feature makes it difcult for medical physicists to assay sources preceding clinical implantation. The AAPM has issued preliminary guidance on this topic,162,163 and is in the process of preparing a report updating prior AAPM recommendations on medical physicist responsibilities164,165 and source assay criteria.166 HDR 192Ir for prostate brachytherapy has been a treatment choice for two decades, with a role primarily for large glands.167,168 A recent possible alternative is HDR 169Yb, which has lower photon energies than 192Ir and a potentially more advantageous dose distribution for localized treatment.98,169 Similarly, LDR 131Cs sources, commercially available since 2004, emit photon energies slightly higher than those of 125I.170,171 Due to the different dose distributions because of photon energy for HDR 169Yb versus HDR 192 Ir, and due to different half-lives for LDR 131Cs versus LDR 125I, there will be different biological responses with these new radiotherapy modalities. Part of the physicists job is to know the science supporting radiation therapy treatment modalities, and to guide the radiation oncologist towards meaningful programmatic decisions regarding choice of treatment modalities.12 This ethical mandate becomes clouded when referral patterns rather than evidence-based medicine guide treatment modality choice, such as has been

4717

Thomadsen et al.: Brachy Anniversary Paper

4717

observed for prostate cryotherapy and radiofrequency ablation.172174 Signicant long-term advances for prostate cancer therapy will come only through clinical results obtained via randomized trials without physician nancial conicts. Once in the operating room OR, after the treatment modality has been decided, there are still many choices on how to proceed. Surprisingly, there is still ambiguity regarding the necessary margin to apply for adequate prostate coverage, even though a collaboration of academically oriented prostate brachytherapy teams endeavored to resolve this issue.175,176 Additional areas under investigation or development are intra-operative treatment planning or IORT where knowledge obtained about seed placement in the OR guides decisions about subsequent seed placement to obtain an optimal plan.15 As medicine becomes more automated, robotic seed implantation may have a signicant role in the future.177179 This technique could potentially improve standardization of the actual seed implantation process and minimize the impact of physician-to-physician technique variations. As in the case of treatment margins, there is no rm consensus on the post-implant time that provides the most representative patient anatomy for which to determine dose for LDR implants. Yue and colleagues have studied this problem with various biomechanical assumptions of prostate edema,180 Tanaka and colleagues performed a 74 patient prospective study correlating need for post-edema imaging with implant quality,181 and AAPM recommendations currently under review have proposed to widen the current reporting criteria towards adding statistical analysis to retrospective studies to address this dilemma.182 On almost all fronts, the eld of prostate brachytherapy has made signicant advances in the past decade. Only the future will reveal which of these technological advances and methodological improvements will become the standard of care for the next generation of medical physicists.

III.C. Breast brachytherapy

One of the fastest growing medical procedures is breast brachytherapy. While in the 1970s and 1980s LDR iridium interstitial implants of the breast were sometimes used to boost the dose to the site of the tumor following whole breast irradiation, this most recent wave of applications uses brachytherapy to implement 1 week long accelerated partial breast irradiation as an alternative to 6 weeks of wholebreast external beam radiation therapy. In the early 1990s, Kuske and colleagues developed techniques for brachytherapy implants following tylectomy segmental mastectomy and subsequently lumpectomy as the sole radiotherapy.183 Initially, the implants consisted of two planes of catheters laid in place during the surgical procedure which did not produce good implant geometries upon closing the operative site, but soon the implants were performed after healing of the surgical site, guided by digital mammograms, ultrasound, or computer tomography.184 Many approaches and techniques, all image-guided, have been develMedical Physics, Vol. 35, No. 10, October 2008

oped and rened since the beginning.185 Most techniques can be learned with relatively short apprenticeships. The MammoSite originally proxima, currently Hologic, Bedford, MA was designed to be a simpler alternative to the interstitial implants, replacing the insertion and treatment of many catheters with a single catheter centered in a balloon that lls the lumpectomy cavity.7,186 With the simplicity of the procedure, many surgeons perform the insertion, which helps bring them into the treatment team, increasing the number of cases treated. The trade-off for the simplicity is a sacrice of control over the dose distribution. For these intracavitary treatments, the prescribed dose contour falls 1 cm outside of the balloon surface. For spherical balloons, a single dwell position in the center serves as the source. Due to the anisotropic dose distribution of the HDR source, the single dwell position results in low doses near the source axis, leading some practitioners to add lightly weighted dwell positions near where the catheter crosses the balloon surface. Oblong balloons require multiple dwell positions. The dose decreases continually with distance from the source center, so the dose near the surface of the balloon tends to be 1.7 to 2.25 times the prescription dose. Skin doses will exceed the prescription dose if the balloon surface lies closer than 1 cm to the skin, leading to a recommendation of a minimal separation between the balloon and the skin of 6 mm. One common problem with these applications occurs when air becomes trapped on the surface of the balloon during insertion, pushing target tissue away from the prescribed dose contour. In attempts to combine the simplicity of the single catheter insertion and the ability of the multi-catheter implants to tailor the dose distribution to the target, a new set of applicators were developed that contained several catheters usually about six connected at the tip and back, bunched together for insertion into the cavity, that then spread into a more spherical shape to ll the cavity. One device, SAVI Cianna Medical, Aliso Viejo, CA, uses the same catheters to spread the cavity and provide paths for the source. ClearPath North American Scientic, Chatsworth, CA has an outer set of catheters that spread the cavity and an inner set for source travel. A hybrid design, Contura SenoRx, Alsio Viejo, CA, has several catheters inside of a balloon. In concept, having multiple catheters in the breast cavity allows reducing the dose in some directions, such as toward the skin, while continuing to push the dose to the prescription distance in other directions, covering the target with increased uniformity. Those systems without the balloon will need to address the dosimetric effects of air within the cavity. Electronic brachytherapy EB, using x-ray units for the radiation source, forms another interesting development. EB comes in two manifestations: the InterBeam system, Carl Zeiss, Oberkochen, Germany, which shoots an electron beam down a long tube to a target centered in an applicator, and the Axxent system Xoft Inc., Fremont, CA, a miniature x-ray tube that moves through a catheter in a manner similar to a conventional, radionuclide high dose-rate unit. These units generate 50 kVp x rays, with considerably less penetration than a 192Ir source. Thus, the dose at the surface

4718

Thomadsen et al.: Brachy Anniversary Paper

4718

of the balloon and the effects of heterogeneities will be higher with EB than with conventional HDR sources. Currently, the EB units cannot treat interstitial type implants. Should they evolve to acquire this ability, these disadvantages would be of less concern. Given the rapid proliferation of partial breast irradiation techniques and devices over the last decade, rapid development of techniques and procedures is likely to continue until a smaller set of alternatives surface as the preferred method. A national trial comparing whole breast irradiation to partial breast irradiation National Surgical Adjuvant Breast and Bowel Project B-39/Radiation Therapy Oncology Group 0413, delivered either as interstitial, intracavitary, or intensity-modulated external-beam irradiation, is unlikely to provide evidence of the superiority of one technique over another. This is in part due to the inclusiveness of the study, allowing centers to enter patients even though they could not randomize the patient to any of the arms, but also because that is not the question the study intends to answer: Does partial breast irradiation, however implemented, yield clinical outcomes as good as or better than that of whole breast irradiation?
III.D. Lung brachytherapy

Lung cancer is one of the leading causes of cancer death for both men and women in the United States.187 Photodynamic therapy PDT,188 brachytherapy,189,190 191 192 electrocautery, cryotherapy, and Nd-YAG laser therapy193 are therapeutic options available for management of endobronchial malignancies. In cases of the tumor being in the lung tissue, rather than the bronchi, external beam radiation therapy is the appropriate therapy.194 All of these treatment modalities have been used for both palliation of late obstructing cancers and primary treatment of early radiographically occult cancers. The signicance of brachytherapy treatment for endobronchial cancer has been demonstrated by many investigators.195200 Intraluminal bronchial irradiation is used to give an added radiation dose to lung cancers that are located in the major bronchi the breathing passages. While increasing the dose to the tumor, brachytherapy, due to its short treatment distance, minimizes the radiation dose given to sensitive nearby tissues such as adjacent normal lung, heart, esophagus and the spinal cord. Brachytherapy is also used to treat recurrent endobronchial tumors where additional surgery and external beam radiation therapy are no longer options. Conventionally, LDR brachytherapy uses 192Ir ribbons. For these treatments, the brachytherapy sources were placed in catheters introduced in the patients bronchus through the nostril and located near the tumor site. Placement of the catheters is normally performed using a bronchoscope, allowing visualization of the exact location of the tumor. In addition, the procedures are performed under uoroscopy to identify a landmark for determination of the treatment length. Often, two images are taken during the procedure, one with the tip of the bronchoscope at the distal margin of the tumor and
Medical Physics, Vol. 35, No. 10, October 2008

one at the proximal margin. The treatment length is determined by adding at least 1 cm to each end of the marked tumor length. These catheters must be carefully secured to the patients nose to prevent dislodgement during the treatment. In this treatment technique, the patient normally receives a dose of 30 to 40 Gy, prescribed at a 1 cm distance from catheter, in a single implant over 1 to 3 days. However, due to the risk of displacement of the catheter during the treatment and also the possibility of irradiation of surrounding people, these patients require hospitalization and surveillance.201,202 The source activities and arrangement must be custom designed for each patient. Ordering, receiving and handling the brachytherapy sources and their inventory and disposal after the treatment for each patient are among the medical physicists tasks. For these reasons, LDR brachytherapy for the lung has mostly been replaced by HDR techniques. Introduction of HDR brachytherapy eliminated some of the above-noted problems. In HDR treatment technique, catheters will be placed in the patients endobronchial tube, as with the LDR approach. However, unlike the LDR treatment, the HDR treatment is performed in fractionated fashion. The number of fractions and dose per fraction are dependent on the disease stage and also any combination with chemotherapy or external beam therapy. Typically, three to ve fractions of 5 to 7.5 Gy per fraction at 1 cm from the catheter at 1 week time intervals between the fractions, or four twice-daily fractions of 8 Gy would be used.195202 One of the main advantages of the HDR system relative to LDR brachytherapy is the optimization of the dwell times for minimization of dose to normal tissue while the dose to the treatment volume is maximized. While some facilities perform the same optimization using the LDR approach, the HDR treatment planning systems simplify this procedure. In summary, the results of various investigators indicate that the combination of new technology with a better understanding of the biological, clinical, and dosimetric aspects of the radiation holds promise for advancement of this treatment modality. Interstitial implants of the surgical bed during lung surgery have been used off and on since the early days of the 20th century. Recently a clinical trail opened using permanent sources in either a single or double line along the resection.203 The sources are sewn into a mesh and the mesh sewn on the lung. Because the length of the incision is not known before the surgery, treatment planning can only be approximate. Even with the best planning and careful laying of the mesh, upon closing the patient the tissues carrying the sources tend to bunch and the resultant dose may have little relationship to that planned. Since there is evidence of benet from these treatments notwithstanding the dosimetric uncertainty, the problem may be more regulatory than clinical.
III.E. Novel clinical applications and delivery schemes

Brachytherapy has expanded, over the last decade, not only in terms of number of procedures, but in terms of di-

4719

Thomadsen et al.: Brachy Anniversary Paper

4719

versity of clinical applications and techniques as well. This growing clinical diversity is due in part to the advent of 192Ir and 125I sources in the 1970s and 1980s. These new sources allowed implantation of sites, sizes, and geometries that the previous rigid needles would not, while computer- and image-based dose evaluation supported greater dose distribution individualization. High dose-rate units in the 1980s changed the patterns of practice, mainly by adapting older techniques to fractionated, outpatient settings. A much greater change came with IVBT,204 which differed signicantly from conventional brachytherapy: treatment distances were in millimeters, typically shorter than available dosimetric information; source placement was by physicians other than radiation oncologists interventional cardiologists; the target was not cancer; effective and safe doses were poorly understood, as were the target tissues. In addition, biological goal of IVB was unconventional: its goal was not to depopulate a population of malignant tumor cells, but to interrupt a benign proliferative process that compromised efcacy of invasive cardiac catheterization procedures. While some treatments used conventional LDR 192Ir sources within a catheter in the vessel, beta sources were also used. A variety of source congurations proliferated, including radioactive-liquid or -gas lled balloon catheters, catheters with the source material impregnating the wall, and stents incorporating radioactive materials. Dosimetric characteristics and other properties of IVBT sources were carefully examined in the AAPM Task Group No. 60 and No. 149 reports.35,36 The practice of IVBT increased incredibly quickly, passing with great rapidity from research institutions to small hospitals. IVBT work on rening IVBT stopped abruptly, along with the clinical practice, with the availability of drug-eluting stents.205 The diversity of approaches in IVB foreshadowed even more novel delivery schemes and additional applications of brachytherapy, including to other benign clinical conditions. Some of the newer applications appear dissimilar to conventional brachytherapy. Macular degeneration. Brachytherapy for macular degeneration began with 103Pd episcleral plaques similar to the episcleral plaques used for ocular melanoma treatment.206 While this therapy provided benet, a new intraocular approach using 90Sr in a device with a long needle-like tip is now in clinical trials Epi-Rad90 Ophthalmic System, NeoVista, Fremont, CA. The invasive nature of this treatment differs markedly from most previous eye applications, and the treatment using a single small beta source presents extreme dosimetric challenges. Radionuclide-labeled microspheres. Although rst developed in the 1960s,207 the wide-spread use of radiolabeled microspheres followed the commercial availability of the products and approval by the Food and Drug Administration in the early 2000s. The approved use currently is for liver cancer, where the microspheres are injected intra-arterially into the hepatic artery branch that feeds the tumor. The microspheres, being
Medical Physics, Vol. 35, No. 10, October 2008

20 to 40 m in diameter, become lodged in the capillary mouths, irradiating the tumor from the periphery. The radiolabel in the commercial product, 90Y an almost pure beta emitter, penetrates only a few millimeters from the resting place of a sphere. For this treatment, investigators are just beginning to map the distribution of microspheres and establish the dose distribution with any accuracy. Radiolabeled macromolecules. Bordering on systemic therapy, many of the macromolecules are only slightly smaller than microspheres. The mechanism of targeting also differs from conventional systemic therapies, which either were metabolized and absorbed or were loculated in a compartment. Most often, these molecules attach to a target, frequently based on antibody interactions. As with the microspheres, the localization and dose distribution remain investigational as of this writing.208 The last two applications fall into the category of microbrachytherapy, i.e., brachytherapy using microscopic carriers. Sometimes a ne, and often arbitrary, line divides microbrachytherapy from unsealed radionuclide therapy. In microbrachytherapy the radionuclide is tagged to carriers that are relatively large on the microscopic scale, and the physical size of the carrier inuences strongly the localization of the dose distribution. In unsealed radionuclide therapy, the source carrier tends toward smaller molecules that often localize through metabolic processes, although, as noted above, these differentiations are only general. Very likely, the use and development of microbrachytherapy will increase in the near future, and the issues surrounding the dose distribution will be resolved. IV. CONCLUSION There has not been a period since its beginning that brachytherapy has not been a dynamic and changing discipline. The current era presents a face of brachytherapy that is changing faster than ever, with a future that portends an even greater diversity of sources and applications. The AAPM has been at the forefront of brachytherapy development, particularly with the work of task groups under the Brachytherapy Subcommittee.
Author to whom correspondence should be addressed. Electronic mail: Thomadsen@humon.wisc.edu. Telephone: 608-263-4183. 1 J. F. Williamson, Brachytherapy technology and physics practice since 1950: a half-century of progress, Phys. Med. Biol. 5113, R303R325 2006. 2 G. S. Ibbott et al., Fifty years of AAPM involvement in radiation dosimetry, Med. Phys. 35, 14181427 2008. 3 J. F. Williamson and M. J. Rivard, Quantitative Dosimetry Methods for Brachytherapy, in Brachytherapy Physics, 2nd ed., edited by B. R. Thomadsen, M. J. Rivard, and W. M. Butler Medical Physics, Madison, WI, 2005, pp. 233294. 4 P. Grimm and J. E. Sylvester, Advances in Brachytherapy, Rev. Neurol. 6 Supp. 4, 3748 2004. 5 L. W. Cuttino, D. Todor, and D. W. Arthur, CT-guided multi-catheter insertion technique for partial breast brachytherapy: Reliable target coverage and dose homogeneity, Brachytherapy 41, 1017 2005. 6 F. A. Vicini et al., Accelerated treatment of breast cancer, J. Clin. Oncol. 197, 19932001 2001.
a

4720
7

Thomadsen et al.: Brachy Anniversary Paper

4720 of brachytherapy nuclides, with particular reference to iodine-125, Med. Phys. 102, 176183 1983. 32 R. Dale, Revisions to radial dose function data for 125I and 131Cs, Med. Phys. 136, 963964 1986. 33 L. L. Anderson, R. Nath, and K. A. Weaver, Interstitial Collaborative Working Group (ICWG). Interstitial Brachytherapy: Physical, Biological, and Clinical Considerations Raven, New York, 1990. 34 R. Nath et al., Dosimetry of interstitial brachytherapy sources: recommendations of the AAPM Radiation Therapy Committee Task Group No. 43. American Association of Physicists in Medicine, Med. Phys. 222, 209234 1995. 35 R. Nath et al., Intravascular brachytherapy physics: report of the AAPM Radiation Therapy Committee Task Group no. 60. American Association of Physicists in Medicine, Med. Phys. 262, 119152 1999. 36 S. T. Chiu-Tsao et al., Dose calculation formalisms and consensus dosimetry parameters for intravascular brachytherapy dosimetry: Recommendations of the AAPM Therapy Physics Committee Task Group No. 149, Med. Phys. 3411, 41264157 2007. 37 A. S. Meigooni et al., Treatment planning consideration for prostate implants with the new linear RadioCoil 103Pd brachytherapy source, J. Appl. Clin. Med. Phys. 63, 2336 2005. 38 L. Liu, S. C. Prasad, and D. A. Bassano, Determination of Cs-137 dosimetry parameters according to the AAPM TG-43 formalism, Med. Phys. 31, 477483 2004. 39 R. Nath et al., Specication of Brachytherapy Source Strength: Report of AAPM Task Group No. 32 American Institute of Physics, Melville, NY, 1987. 40 K. E. Stump, L. A. DeWerd, J. A. Micka, and D. R. Anderson Calibration of new high dose rate 192Ir sources, Med. Phys. 297, 14831488 2002. 41 B. Rasmussen, S. Davis, J. Micka, and L. DeWerd, The Air-Kerma Strength Standard for 192Ir HDR Sources at the University of Wisconsin ADCL, Med. Phys. 356, 2969 2008. 42 R. Colle and B. E. Zimmerman, A dual-compensated cryogenic microcalorimeter for radioactivity standardizations, Appl. Radiat. Isot. 5612, 223230 2002. 43 A. Sarfehnia, K. Stewart, and J. Seuntjens, An absorbed dose to water standard for HDR 192Ir brachytherapy sources based on water calorimetry: Numerical and experimental proof-of-principle, Med. Phys. 3412, 49574961 2007. 44 Z. Chen and R. Nath, Dose rate constants determined by a photon spectrometry technique for 20 different models of low-energy brachytherapy sources, Med. Phys. 35, 2864 2008. 45 A. S. Meigooni et al., Instrumentation and dosimeter-size artifacts in quantitative thermoluminescence dosimetry of low-dose elds, Med. Phys. 225, 555561 1995. 46 J. F. Williamson and A. S. Meigooni, Quantitative Dosimetry Methods in Brachytherapy, in Brachytherapy Physics, edited by J. F. Williamson, B. R. Thomadsen, and R. Nath Medical Physics, Madison, WI, 1995. 47 E. Y. Hirata et al., Low dose fraction behavior of high sensitivity radiochromic lm, Med. Phys. 324, 10541060 2005. 48 S. T. Chiu-Tsao et al., Dose response characteristics of new models of GAFCHROMIC lms: Dependence on densitometer light source and radiation energy, Med. Phys. 319, 25012508 2004. 49 S. T. Chiu-Tsao et al., Verication of Ir-192 near source dosimetry using GAFCHROMIC lm, Med. Phys. 312, 201207 2004. 50 S. T. Chiu-Tsao et al., Energy dependence of response of new high sensitivity radiochromic lms for megavoltage and kilovoltage radiation energies, Med. Phys. 3211, 33503354 2005. 51 K. A. Gifford et al., Comparison of Monte Carlo calculations around a Fletcher Suit Delclos ovoid with radiochromic lm and normoxic polymer gel dosimetry, Med. Phys. 327, 22882294 2005. 52 P. Baras et al., An evaluation of the TSE MR sequence for time efcient data acquisition in polymer gel dosimetry of applications involving high doses and steep dose gradients, Med. Phys. 3211, 33393345 2005. 53 E. Pantelis et al., Polymer gel dosimetry close to an 125I interstitial brachytherapy seed, Phys. Med. Biol. 5018, 43714384 2005. 54 P. Papagiannis et al., Polymer gel dosimetry for the TG-43 dosimetric characterization of a new 125I interstitial brachytherapy seed, Phys. Med. Biol. 518, 21012111 2006. 55 R. G. Selwyn, Image-based dosimetry for selective internal radiation therapy SIRT using Y-90 microspheres, University of Wisconsin, 2007. 56 R. M. Sievert, Die Intensitatsverteilung der primaren g-strahlung in der

M. Keisch et al., Initial clinical experience with the MammoSite breast brachytherapy applicator in women with early-stage breast cancer treated with breast-conserving therapy, Int. J. Radiat. Oncol., Biol., Phys. 552, 289293 2003. 8 M. B. Leon et al., Localized intracoronary gamma-radiation therapy to inhibit the recurrence of restenosis after stenting, N. Engl. J. Med. 3444, 250256 2001. 9 R. Waksman, Vascular brachytherapy: Applications in the era of drugeluting stents, Rev Cardiovasc Med 3 Suppl. 5, S23S30 2002. 10 G. W. Stone et al., Paclitaxel-eluting stents vs vascular brachytherapy for in-stent restenosis within bare-metal stents: The TAXUS V ISR randomized trial, JAMA, J. Am. Med. Assoc. 29511, 12531263 2006. 11 J. W. Moses et al., Sirolimus-eluting stents versus standard stents in patients with stenosis in a native coronary artery, N. Engl. J. Med. 349, 13151323 2003. 12 Y. Yu et al., Permanent prostate seed implant brachytherapy: Report of the American Association of Physicists in Medicine Task Group No. 64, Med. Phys. 2610, 20542076 1999. 13 R. K. Das et al., 3D CT-based high-dose-rate breast brachytherapy implants: Treatment planning and quality assurance, Int. J. Radiat. Oncol., Biol., Phys. 594, 12241228 2004. 14 C. Haie-Meder et al., Recommendations from Gynaecological GYN GEC-ESTRO Working Group I: concepts and terms in 3D image based 3D treatment planning in cervix cancer brachytherapy with emphasis on MRI assessment of GTV and CTV, Radiother. Oncol. 743, 235245 2005. 15 S. Nag et al., Intraoperative planning and evaluation of permanent prostate brachytherapy: Report of the American Brachytherapy Society, Int. J. Radiat. Oncol., Biol., Phys. 515, 14221430 2001. 16 E. K. Lee and M. Zaider, Intraoperative dynamic dose optimization in permanent prostate implants, Int. J. Radiat. Oncol., Biol., Phys. 563, 854861 2003. 17 T. Martin et al., 3-D conformal HDR brachytherapy as monotherapy for localized prostate cancer. A pilot study, Strahlenther. Onkol. 1804, 225232 2004. 18 R. S. Malyapa et al., Physiologic FDG-PET Three-Dimensional Brachytherapy Treatment Planning for Cervical Cancer, Int. J. Radiat. Oncol., Biol., Phys. 544, 11401146 2002. 19 M. J. Rivard et al., Update of AAPM Task Group No. 43 Report: A revised AAPM protocol for brachytherapy dose calculations, Med. Phys. 313, 633674 2004. 20 O. Chibani and J. F. Williamson, MCPI: a sub-minute Monte Carlo dose calculation engine for prostate implants, Med. Phys. 3212, 36883698 2005. 21 A. K. Tedgren and A. Ahnesjo, Accounting for high Z shields in brachytherapy using collapsed cone superposition for scatter dose calculation, Med. Phys. 308, 22062217 2003. 22 J. F. Williamson, Integration of IMRT and Brachytherapy, in IMRT Handbook, edited by T. Bortfeld, R. Schmidt-Ullrich, W. DeNeve et al. Springer, Heidelberg, 2005, pp. 423438. 23 M. Van Herk, Errors and margins in radiotherapy, Semin. Radiat. Oncol. 141, 5264 2004. 24 M. van Herk, Different styles of image-guided radiotherapy, Semin. Radiat. Oncol. 174, 258267 2007. 25 R. Timmerman et al., Optimizing dose and fractionation for stereotactic body radiation therapy. Normal tissue and tumor control effects with large dose per fraction, Front. Radiat. Ther. Oncol. 40, 352365 2007. 26 T. Pawlicki, C. Cotrutz, and C. King, Prostate cancer therapy with stereotactic body radiation therapy, Front. Radiat. Ther. Oncol. 40, 395406 2007. 27 L. Xiong et al., Deformable structure registration of bladder through surface mapping, Med. Phys. 336, 18481856 2006. 28 G. E. Christensen et al., Image-based dose planning of intracavitary brachytherapy: Registration of serial-imaging studies using deformable anatomic templates, Int. J. Radiat. Oncol., Biol., Phys. 511, 227243 2001. 29 M. B. Sharpe and K. Brock, Quality assurance of serial 3D image registration, fusion and segmentation, Int. J. Radiat. Oncol., Biol., Phys., Suppl. 71 1 Suppl., S33S37 2008. 30 L. L. Meisberger, R. J. Keller, and R. J. Shalek, The effective attenuation in water of the gamma rays of gold-198, iridium-192, cesium-137, radium-226, and cobalt-60, Radiology 905, 953957 1968. 31 R. G. Dale, Some theoretical derivations relating to the tissue dosimetry Medical Physics, Vol. 35, No. 10, October 2008

4721

Thomadsen et al.: Brachy Anniversary Paper

4721 cross-sections and tally methods for low-energy photon emitters, Phys. Med. Biol. 478, 13211332 2002. 85 D. E. Cullen, J. H. Hubbell, and L. Kissel, EPDL97: The evaluated photon data library, 97 Version, Vol. 6, Revision 5, Report No. UCRL50400, Lawrence Livermore National Laboratory, 1997. 86 J. Perez-Calatayud, D. Granero, and F. Ballester, Phantom size in brachytherapy source dosimetric studies, Med. Phys. 317, 20752081 2004. 87 G. Lymperopoulou et al., A dosimetric comparison of 169Yb and 192Ir for HDR brachytherapy of the breast, accounting for the effect of nite patient dimensions and tissue inhomogeneities, Med. Phys. 3312, 4583 4589 2006. 88 R. E. Taylor, G. Yegin, and D. W. Rogers, Benchmarking brachydose: Voxel based EGSnrc Monte Carlo calculations of TG-43 dosimetry parameters, Med. Phys. 342, 445457 2007. 89 A. S. Meigooni, J. A. Meli, and R. Nath, Interseed effects on dose for 125I brachytherapy implants, Med. Phys. 192, 385390 1992. 90 O. Chibani, J. F. Williamson, and D. Todor, Dosimetric effects of seed anisotropy and interseed attenuation for 103Pd and 125I prostate implants, Med. Phys. 328, 25572566 2005. 91 M. J. Rivard, Monte Carlo calculations of AAPM Task Group Report No. 43 dosimetry parameters for the MED3631-A/M 125I source, Med. Phys. 28, 629637 2001. 92 S. M. Seltzer et al., New national air-kerma-strength standards for 125I and 103Pd brachytherapy seeds, J. Res. Natl. Inst. Stand. Technol. 108, 337358 2003. 93 J. F. Williamson, Dosimetric characteristics of the DRAXIMAGE model LS-1 1-125 interstitial brachytherapy source design: A Monte Carlo investigation, Med. Phys. 294, 509-521 2002. 94 D. A. Low et al., Applicator-guided intensity-modulated radiation therapy, Int. J. Radiat. Oncol., Biol., Phys. 525, 14001406 2002. 95 C. S. Melhus and M. J. Rivard, Approaches to calculating AAPM TG-43 brachytherapy dosimetry parameters for 137Cs, 125I, 192Ir, 103Pd, and 169Yb sources, Med. Phys. 336, 17291737 2006. 96 M. J. Rivard et al., Calculated and measured brachytherapy dosimetry parameters in water for the Xoft Axxent X-Ray Source: an electronic brachytherapy source, Med. Phys. 3311, 40204032 2006. 97 M. C. Smitt and R. Kirby, Dose-volume characteristics of a 50-kV electronic brachytherapy source for intracavitary accelerated partial breast irradiation, Brachytherapy 63, 207211 2007. 98 D. C. Medich, M. A. Tries, and J. J. Munro, 2nd, Monte Carlo characterization of an ytterbium-169 high dose rate brachytherapy source with analysis of statistical uncertainty, Med. Phys. 331, 163172 2006. 99 G. Yegin and D. W. O. Rogers A fast Monte Carlo code for multi-seed brachytherapy treatments including interseed effects, Med. Phys. 31, 1771 2004. 100 A. K. Carlsson and A. Ahnesjo, The collapsed cone superposition algorithm applied to scatter dose calculations in brachytherapy, Med. Phys. 2710, 23202332 2000. 101 K. R. Russell, A. K. Tedgren, and A. Ahnesjo, Brachytherapy source characterization for improved dose calculations using primary and scatter dose separation, Med. Phys. 329, 27392752 2005. 102 C. Boudreau et al., IMRT head and neck treatment planning with a commercially available Monte Carlo based planning system, Phys. Med. Biol. 505, 879890 2005. 103 K. A. Gifford et al., A three-dimensional computed tomography-assisted Monte Carlo evaluation of ovoid shielding on the dose to the bladder and rectum in intracavitary radiotherapy for cervical cancer, Int. J. Radiat. Oncol., Biol., Phys. 632, 615621 2005. 104 J. Markman et al., On the validity of the superposition principle in dose calculations for intracavitary implants with shielded vaginal colpostats, Med. Phys. 282, 147155 2001. 105 A. S. Kirov et al., Measurement and calculation of heterogeneity correction factors for an Ir-192 high dose-rate brachytherapy source behind tungsten alloy and steel shields, Med. Phys. 236, 911919 1996. 106 H. Perera et al., Dosimetric Characteristics, Air-kerma Strength Calibration and Verication of Monte Carlo Simulation for a New Ytterbium-169 Brachytherapy Source, Int. J. Radiat. Oncol., Biol., Phys. 28, 953971 1994. 107 J. F. Williamson, Dose calculations about shielded gynecological colpostats, Int. J. Radiat. Oncol., Biol., Phys. 19, 167178 1990. 108 J. F. Williamson, Semi-empirical Dose-Calculation Models in Brachytherapy, in Brachytherapy Physics, 2nd ed., edited by B. R. Thomadsen,

nahe medizinischer radiumpraparate, Acta Radiol. 1921-1962 1, 89 128 1921. 57 M. A. Van Dilla and G. J. Hine, Gamma-ray diffusion experiments in water, Nucleonics 10, 5458 1952. 58 R. J. Shalek and M. Stovall, The M. D. Anderson method for the computation of isodose curves around interstitial and intracavitary radiation sources. I. Dose from linear sources, Am. J. Roentgenol., Radium Ther. Nucl. Med. 1023, 662672 1968. 59 V. Krishnaswamy, Calculation of the dose distribution about californium-252 needles in tissue, Radiology 981, 155160 1971. 60 G. E. Moore, Cramming more components onto integrated circuits, Electronics 38, 114117 1965. 61 M. J. Rivard et al., Clinical brachytherapy with neutron emitting 252Cf sources and adherence to AAPM TG-43 dosimetry protocol, Med. Phys. 261, 8796 1999. 62 M. J. Berger, MIRD Pamphlet 2: Energy deposition in water by photons from point isotropic sources, J. Nucl. Med. 9 Supp. 1, 1725 1968. 63 S. Webb and R. A. Fox, The dose in water surrounding point isotropic gamma-ray emitters, Br. J. Radiol. 52618, 482484 1979. 64 J. F. Williamson, R. L. Morin, and F. M. Khan, Monte Carlo evaluation of the Sievert integral for brachytherapy dosimetry, Phys. Med. Biol. 289, 10211032 1983. 65 G. S. Burns and D. E. Raeside, Monte Carlo simulation of the dose distribution around 125I seeds, Med. Phys. 143, 420424 1987. 66 J. F. Williamson and T. Seminoff, Template-guided interstitial implants: Cs-137 reusable sources as a substitute for Ir-192, Radiology 1651, 265269 1987. 67 A. S. Meigooni, J. A. Meli, and R. Nath, A comparison of solid phantoms with water for dosimetry of 125I brachytherapy sources, Med. Phys. 155, 695701 1988. 68 J. F. Williamson, Monte Carlo and analytic calculation of absorbed dose near 137Cs intracavitary sources, Int. J. Radiat. Oncol., Biol., Phys. 151, 227237 1988. 69 J. F. Williamson, Monte Carlo evaluation of specic dose constants in water for 125I seeds, Med. Phys. 155, 686694 1988. 70 J. F. Williamson and F. J. Quintero, Theoretical evaluation of dose distributions in water about models 6711 and 6702 125I seeds, Med. Phys. 156, 891897 1988. 71 J. F. Williamson, R. L. Morin, and F. M. Khan, Dose calibrator response to brachytherapy sources: A Monte Carlo and analytic evaluation, Med. Phys. 102, 135140 1983. 72 W. H. Ellett, G. L. Brownell, and A. R. Reddy, An assessment of Monte Carlo calculations to determine gamma ray dose from internal emitters, Phys. Med. Biol. 132, 219230 1968. 73 J. F. Williamson et al., Comparison of calculated and measured heterogeneity correction factors for 125I, 137Cs, and 192Ir brachytherapy sources near localized heterogeneities, Med. Phys. 201, 209222 1993. 74 G. W. Batten, The M. D. Anderson method for the computation of isodose curves around interstitial and intracavitary radiation sources: II. Mathematical and computational aspect, Am. J. Roentgenol. 102, 673 676 1968. 75 M. L. Meurk and G. D. Adams, Computer evaluation of dose rates around multiple radium sources, Radiology 80, 115116 1963. 76 G. D. Adams and M. L. Meurk, The use of a computer to calculate isodose information surrounding distributed gynaecological radium sources, Phys. Med. Biol. 18, 533540 1964. 77 R. F. Nelson and M. L. Meurk, Use of automatic computing machines for implant dosimetry, Radiology 70, 90 1958. 78 C. S. Hope et al., Computerization of dose distribution in cervix radium treatments, Phys. Med. Biol. 9, 345390 1964. 79 R. J. Shalek and M. A. Stovall, Calculation of isodose distributions in interstitial implantations by computer, Radiology 76, 119120 1961. 80 M. A. Stovall and R. J. Shalek, Study of explicit distributions of radiation in interstitial implantations. I. Method of calculation with automatic digital computer, Radiology 78, 950954 1962. 81 J. F. Williamson, Monte Carlo evaluation of kerma at a point for photon transport problems, Med. Phys. 144, 567576 1987. 82 J. G. Wierzbicki et al., Calculated dosimetric parameters of the IoGold 125 I source model 3631-A, Med. Phys. 2511, 21972199 1998. 83 S. E. Storm and H. I. Israel, Photon cross-sections from 1 keV to 100 MeV for elements Z = 1 to Z = 100, Nucl. Data, Sect. A 7, 566581 1970. 84 J. J. Demarco, R. E. Wallace, and K. Boedeker, An analysis of MCNP Medical Physics, Vol. 35, No. 10, October 2008

4722

Thomadsen et al.: Brachy Anniversary Paper

4722 aspects, Br. J. Radiol. 11, 252266 1938. D. Neblett, Techniques and Applicators Available for Interstitial Implantation, in Brachytherapy Physics, edited by J. F. Williamson, B. R. Thomadsen, and R. Nath Medical Physics, Madison, WI, 1995. 137 B. R. Thomadsen, S. Shahabi, and D. A. Buchler, Differential loadings of brachytherapy templates, Endocurietherapy/Hyperthermia Oncology 6, 197202 1990. 138 G. Ezzell, Optimization in Brachytherapy, in Brachytherapy Physics, 2nd ed., edited by B. R. Thomadsen, M. J. Rivard, and W. Butler Medical Physics, Madison, WI, 2005. 139 G. Ezzell and R. W. Luthmann, Clinical Implementation of Dwell Time Optimization Techniques, in Brachytherapy Physics, edited by J. F. Williamson, B. R. Thomadsen, and R. Nath Medical Physics, Madison, WI, 1995. 140 E. K. Lee et al., Treatment planning for brachytherapy: an integer programming model, two computational approaches and experiments with permanent prostate implant planning, Phys. Med. Biol. 441, 145165 1999. 141 W. D. DSouza et al., An iterative sequential mixed-integer approach to automated prostate brachytherapy treatment plan optimization, Phys. Med. Biol. 462, 297322 2001. 142 J. Pouliot, E. Lessard, and I.-C. Hsu, Advanced 3D Planning, in Brachytherapy Physics, 2nd ed., edited by B. R. Thomadsen, M. J. Rivard, and W. Butler Medical Physics, Madison, WI, 2005. 143 S. Yoo et al., A greedy heuristic using adjoint functions for the optimization of seed and needle congurations in prostate seed implant, Phys. Med. Biol. 523, 815828 2007. 144 B. R. Thomadsen, Achieving Quality in Brachytherapy Taylor and Francis, London, 1999. 145 M. A. Cleaves, Radium: With a preliminary note on radium rays in the treatment of cancer, Medical Record 6416, 601606 1903. 146 G. H. Fletcher, Uterine Cervix, in Textbook of Radiotherapy, edited by G. H. Fletcher Lea & Febiger, Philadelphia, 1966, pp. 434474. 147 O. Pasteau and P. Degrais, The radium treatment of cancer of the prostate, Arch Roentgenol Ray 18, 396410 1914. 148 W. M. Butler and G. S. Merrick, Introduction to prostate brachytherapy, in Brachytherapy Physics, 2nd ed., edited by B. R. Thomadsen, M. J. Rivard, and W. Butler Medical Physics, Madison, WI, 2005. 149 W. M. Butler and G. S. Merrick, Treatment planning in permanent prostate brachytherapy, in Brachytherapy Physics, 2nd ed., edited by B. R. Thomadsen, M. J. Rivard, and W. M. Butler Medical Physics, Madison, WI, 2005. 150 E. P. Lief, Treatment Delivery in Prostate LDR Brachytherapy, in Brachytherapy Physics, 2nd ed., edited by B. R. Thomadsen, M. J. Rivard, and W. M. Butler Medical Physics, Madison, WI, 2005, pp. 589 601. 151 W. S. Bice, Post procedural evaluation for prostate implants, in Brachytherapy Physics, 2nd ed., edited by B. R. Thomadsen, M. J. Rivard, and W. M. Butler Medical Physics, Madison, WI, 2005, pp. 603640. 152 E. P. Lief, Modern advances in prostate brachytherapy, in Brachytherapy Physics, 2nd ed., edited by B. R. Thomadsen, M. J. Rivard, and W. M. Butler Medical Physics, Madison, WI, 2005, pp. 657672. 153 Z. Ouhib, HDR brachytherapy for prostate, in Brachytherapy Physics, 2nd ed., edited by B. R. Thomadsen, M. J. Rivard, and W. M. Butler Medical Physics, Madison, WI, 2005, pp. 641656. 154 Z. Wang and N. E. Hertel, Determination of dosimetric characteristics of OptiSeedTM a plastic brachytherapy 103Pd source, Appl. Radiat. Isot. 633, 311321 2005. 155 S. Bernard and S. Vynckier, Dosimetric study of a new polymer encapsulated palladium-103 seed, Phys. Med. Biol. 507, 14931504 2005. 156 A. S. Meigooni et al., Dosimetric characteristics of the new RadioCoil 103 Pd wire line source for use in permanent brachytherapy implants, Med. Phys. 3111, 30953105 2004. 157 A. S. Meigooni, S. B. Awan, and K. Dou, Feasibility of calibrating elongated brachytherapy sources using a well-type ionization chamber, Med. Phys. 3311, 41844189 2006. 158 A. B. Paxton et al., Primary calibration of coiled 103Pd brachytherapy sources, Med. Phys. 351, 3238 2008. 159 D. C. Medich and J. J. Munro, 3rd, Monte Carlo calculated TG-43 dosimetry parameters for the SeedLink 125Iodine brachytherapy system, Med. Phys. 309, 25032508 2003. 160 L. Lin et al., The use of directional interstitial sources to improve dosimetry in breast brachytherapy, Med. Phys. 351, 240247 2008.
136

M. J. Rivard, and W. M. Butler Medical Physics, Madison, WI, 2005, pp. 201232. 109 P. Lindsay, J. Battista, and J. Van Dyk, The effect of seed anisotropy on brachytherapy dose distributions using 125I and 103Pd, Med. Phys. 283, 336345 2001. 110 D. Y. Huang et al., Dose distribution of 125I sources in different tissues, Med. Phys. 175, 826832 1990. 111 A. S. Meigooni and R. Nath, Tissue inhomogeneity correction for brachytherapy sources in a heterogeneous phantom with cylindrical symmetry, Med. Phys. 192, 401407 1990. 112 R. K. Das et al., Validation of Monte Carlo dose calculations near 125I sources in the presence of bounded heterogeneities, Int. J. Radiat. Oncol., Biol., Phys. 384, 843853 1997. 113 R. L. Maughan et al., The elemental composition of tumors: Kerma data for neutrons, Med. Phys. 248, 12411244 1997. 114 G. Anagnostopoulos et al., The effect of patient inhomogeneities in oesophageal 192Ir HDR brachytherapy: A Monte Carlo and analytical dosimetry study, Phys. Med. Biol. 4912, 26752685 2004. 115 S. J. Ye et al., Dose errors due to inhomogeneities in balloon catheter brachytherapy for breast cancer, Int. J. Radiat. Oncol., Biol., Phys. 602, 672677 2004. 116 J. F. Williamson, R. Baker, and Z. Li, A convolution algorithm for brachytherapy dose computations in heterogeneous geometries, Med. Phys. 18, 12561265 1991. 117 G. M. Daskalov et al., Dosimetric modeling of the microselectron highdose rate 192Ir source by the multigroup discrete ordinates method, Med. Phys. 2710, 23072319 2000. 118 G. M. Daskalov et al., Multigroup discrete ordinates modeling of 125I 6702 seed dose distributions using a broad energy-group cross section representation, Med. Phys. 292, 113124 2002. 119 K. A. Gifford et al., Comparison of a nite-element multigroup discreteordinates code with Monte Carlo for radiotherapy calculations, Phys. Med. Biol. 519, 22532265 2006. 120 J. Williamson et al., Accuracy and efciency comparisons between three-dimensional multigroup discrete ordinates and voxel based Monte Carlo methods for dosimetric modeling of the Model 6702 125I seed, Med. Phys. 286, 1229 2001. 121 J. J. DeMarco et al., CT-based dosimetry calculations for 125I prostate implants, Int. J. Radiat. Oncol., Biol., Phys. 455, 13471353 1999. 122 F. A. Lerma and J. F. Williamson, Accurate localization of intracavitary brachytherapy applicators from 3D CT imaging studies, Med. Phys. 293, 325333 2002. 123 ICRP, ICRP Publication 89: Basic Anatomical and Physiological Data for Use in Radiological Protection: Reference Values, International Commission on Radiological Protection Pergamon, Oxford, 2003. 124 C. E. Cann, Quantitative CT for Determination of Bone Mineral Density: A Review, Radiology 166, 509522 1988. 125 R. A. Rutherford, B. R. Pullan, and I. Isherwood, Measurement of effective atomic number and electron density using an EMI scanner, Neuroradiology 111, 1521 1976. 126 S. Devic et al., Dual Energy CT Tissue Quantitation for Monte-Carlo Based Treatment Planning for Brachytherapy, presented at the Proceedings of the 2000 World Congress on Medical Physics and Biomedical Engineering, Chicago, IL, 2000. 127 J. F. Williamson et al., On Two-Parameter Representations of Photon Cross Sections: Application to Dual Energy CT imaging, Med. Phys. 33, 41154129 2006. 128 W. J. Meredith, Radium Dosage E. & S. Livingstone, Edinburgh, 1967. 129 B. Pierquin and G. Marinello, A Practical Manual of Brachytherapy Medical Physics, Madison, WI, 1997. 130 D. K. Kwan et al., Single- and double-plane iridium-192 interstitial implants: implantation guidelines and dosimetry, Med. Phys. 104, 456 461 1983. 131 R. D. Zwicker, R. Schmidt-Ullrich, and B. Schiller, Planning of Ir-192 seed implants for boost irradiation to the breast, Int. J. Radiat. Oncol., Biol., Phys. 1112, 21632170 1985. 132 R. Paterson, A dosage system for gamma ray therapy. Part I: Clinical aspects, Br. J. Radiol. 7, 592612 1934. 133 H. Parker, A dosage system for gamma ray therapy. Part II: Physical aspects, Br. J. Radiol. 7, 612632 1934. 134 H. Parker, A dosage system for gamma ray therapy. Part II: Physical aspects, Br. J. Radiol. 11, 313340 1938. 135 R. Paterson, A dosage system for gamma ray therapy. Part I: Clinical Medical Physics, Vol. 35, No. 10, October 2008

4723
161

Thomadsen et al.: Brachy Anniversary Paper

4723 ogy Williston Park 152, 195202 2001. D. Wazer, D. Arthur, and F. Vincini, Accelerated Partial Breast Irradiation: Techniques and Clinical Implementation Springer, Berlin, 2006. 186 G. K. Edmundson et al., Dosimetric characteristics of the MammoSite RTS, a new breast brachytherapy applicator, Int. J. Radiat. Oncol., Biol., Phys. 524, 11321139 2002. 187 R. T. Greenlee et al., Cancer statistics, 2001, Ca-Cancer J. Clin. 511, 1536 2001. 188 T. Okunaka et al., Photodynamic therapy for multiple primary bronchogenic carcinoma, Cancer 682, 253258 1991. 189 H. Marsiglia et al., High-dose-rate brachytherapy as sole modality for early-stage endobronchial carcinoma, Int. J. Radiat. Oncol., Biol., Phys. 473, 665672 2000. 190 M. Perol et al., Curative irradiation of limited endobronchial carcinomas with high-dose rate brachytherapy. Results of a pilot study, Chest 1115, 14171423 1997. 191 T. Van Boxem et al., Radiographically occult lung cancer treated with beroptic bronchoscopic electrocautery: a pilot study of a simple and inexpensive technique, Eur. Respir. J. 11, 169172 1998. 192 N. Deygas et al., Cryotherapy in early supercial bronchogenic carcinoma, Chest 1201, 2631 2001. 193 S. Cavaliere et al., Nd-YAG laser therapy in lung cancer: an 11-year experience with 2,253 applications in 1,585 patients, J. Bronchol. 1, 105111 1994. 194 C. Aygun et al., Treatment of non-small cell lung cancer with external beam radiotherapy and high dose rate brachytherapy, Int. J. Radiat. Oncol., Biol., Phys. 231, 127132 1992. 195 L. F. Chang et al., High dose rate afterloading intraluminal brachytherapy in malignant airway obstruction of lung cancer, Int. J. Radiat. Oncol., Biol., Phys. 283, 589596 1994. 196 H. Yokomise et al., Long-term remission after brachytherapy with external irradiation for locally advanced lung cancer, Respiration 656, 489491 1998. 197 L. E. Gaspar, Brachytherapy in lung cancer, J. Surg. Oncol. 671, 6070 1998. 198 R. M. Huber et al., Does additional brachytherapy improve the effect of external irradiation? A prospective, randomized study in central lung tumors, Int. J. Radiat. Oncol., Biol., Phys. 383, 533540 1997. 199 D. Ornadel et al., Dening the roles of high dose rate endobronchial brachytherapy and laser resection for recurrent bronchial malignancy, Lung Cancer 1623, 203213 1997. 200 B. L. Speiser and L. Spratling, Remote afterloading brachytherapy for the local control of endobronchial carcinoma, Int. J. Radiat. Oncol., Biol., Phys. 254, 579587 1993. 201 A. G. Villanueva, T. C. Lo, and J. F. Beamis, Jr., Endobronchial brachytherapy, Clin. Chest Med. 163, 445454 1995. 202 B. S. Hilaris, M. S. Porrazzo, and C. R. Moorthy, Endobronchial radiation therapy, in Flexible Bronchoscopy, edited by K. P. Wang and A. C. Metha Blackwell Science, Cambridge, MA, 1995, pp. 275287. 203 American College of Surgeons Oncology Group, A randomized phase III study of sublobar resection versus sublobar resection plus brachytherapy in high risk patients with non-small cell lung cancer NSCLC, 3 cm or smaller, Protocol Z4032 2005. 204 R. Waxman and P. Serruys, Handbook of Vascular Brachytherapy Martin Dunitz, London, 1998. 205 L. M. de la Fuente, J. Mrad, E. Penaloza, A. C. Yeung, R. Eury, M. Froix, P. J. Fitzgerald, and S. H. Stertzer, Initial results of the Quanam drug eluting stent QuaDS-QP-2 Registry BARDDS in human subjects, Catheter Cardiovasc Interv 53, 480488 2001. 206 P. T. Finger et al., Palladium-103 plaque radiation therapy for macular degeneration: Results of a 7 year study, Br. J. Ophthamol. 8712, 1497 1503 2003. 207 J. W. Lafave et al., Y90-tagged microspheres in adjuvant tumor therapy, Surgery St. Louis 53, 778783 1963. 208 G. A. Wiseman et al., Phase I/II 90Y-Zevalin yttrium-90 ibritumomab tiuxetan, IDEC-Y2B8 radioimmunotherapy dosimetry results in relapsed or refractory non-Hodgkins lymphoma, Eur. J. Nucl. Med. 277, 766 777 2000.
185

L. A. DeWerd et al., Calibration of multiple LDR brachytherapy sources, Med. Phys. 3310, 38043813 2006. 162 W. M. Butler et al., Third party brachytherapy seed calibrations and physicist responsibilities, Med. Phys. 331, 247248 2006. 163 M. J. Rivard et al., Third-party brachytherapy source calibrations and physicist responsibilities: Report of the AAPM Low Energy Brachytherapy Source Calibration Working Group, Med. Phys. 359, 3860 3865 2008. 164 G. J. Kutcher et al., Comprehensive QA for radiation oncology: report of AAPM Radiation Therapy Committee Task Group 40, Med. Phys. 214, 581618 1994. 165 R. Nath et al., Code of practice for brachytherapy physics: report of the AAPM Radiation Therapy Committee Task Group No. 56. American Association of Physicists in Medicine, Med. Phys. 2410, 15571598 1997. 166 N. J. Yue, B. G. Haffty, and J. Yue, On the assay of brachytherapy sources, Med. Phys. 346, 19751982 2007. 167 S. J. Damore et al., Needle displacement during HDR brachytherapy in the treatment of prostate cancer, Int. J. Radiat. Oncol., Biol., Phys. 465, 12051211 2000. 168 A. T. Monroe et al., High-dose-rate brachytherapy for large prostate volumes / = 50 cc-Uncompromised dosimetric coverage and acceptable toxicity, Brachytherapy 71, 711 2008. 169 G. Lymperopoulou et al., A dosimetric comparison of 169Yb versus 192Ir for HDR prostate brachytherapy, Med. Phys. 32, 38323842 2005. 170 Z. Chen, P. Bongiorni, and R. Nath, Dose rate constant of a cesium-131 interstitial brachytherapy seed measured by thermoluminescent dosimetry and gamma-ray spectrometry, Med. Phys. 3211, 32793285 2005. 171 M. J. Rivard, Brachytherapy dosimetry parameters calculated for a 131Cs source, Med. Phys. 342, 754762 2007. 172 A. B. Barqawi and E. D. Crawford, The current use and future trends of focal surgical therapy in the management of localized prostate cancer, Cancer 135, 313317 2007. 173 W. C. Huang, C. L. Lee, and J. A. Eastham, Locally ablative therapies for primary radiation failures: a review and critical assessment of the efcacy, Curr Urol Rep 83, 217223 2007. 174 T. J. Wilt et al., Systematic review: comparative effectiveness and harms of treatments for clinically localized prostate cancer, Ann. Intern Med. 1486, 435448 2008. 175 D. R. Reed et al., Effect of post-implant edema on prostate brachytherapy treatment margins, Int. J. Radiat. Oncol., Biol., Phys. 635, 14691473 2005. 176 G. S. Merrick et al., Dosimetry of an extracapsular anulus following permanent prostate brachytherapy, Am. J. Clin. Oncol. 303, 228233 2007. 177 M. J. Rivard, D. A. Evans, and I. Kay, A technical evaluation of the Nucletron FIRST system: conformance of a remote afterloading brachytherapy seed implantation system to manufacturer specications and AAPM Task Group report recommendations, J. Appl. Clin. Med. Phys. 61, 2250 2005. 178 G. Fichtinger et al., Robotic assistance for ultrasound guided prostate brachytherapy, International Conference on Medical Image Computing and Computer Assisted Intervention, Brisbane, Australia, 2007, Vol 10 Pt 1, pp. 119127. 179 M. A. Meltsner, N. J. Ferrier, and B. R. Thomadsen, Observations on rotating needle insertions using a brachytherapy robot, Phys. Med. Biol. 5219, 60276037 2007. 180 N. Yue et al., Prescription dose in permanent 131Cs seed prostate implants, Med. Phys. 328, 24962502 2005. 181 O. Tanaka et al., Effect of edema on postimplant dosimetry in prostate brachytherapy using CT/MRI fusion, Int. J. Radiat. Oncol., Biol., Phys. 692, 614618 2007. 182 R. Nath et al., AAPM recommendations on dose prescription and reporting methods for permanent interstitial brachytherapy for prostate cancer, Med. Phys. in press. 183 T. A. King et al., Long-term results of wide-eld brachytherapy as the sole method of radiation therapy after segmental mastectomy for Tis,1,2 breast cancer, Am. J. Surg. 1804, 299304 2000. 184 S. Nag et al., Brachytherapy in the treatment of breast cancer, Oncol-

Medical Physics, Vol. 35, No. 10, October 2008

También podría gustarte