42
Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder Nath Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut 06510 Lowell L. Anderson Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York 10021 Jerome A. Meli Department of Radiology, St. Vincents Medical Center, Bridgeport, Connecticut 06606 Arthur J. Olch Department of Radiation Oncology, Southern California Permanente Medical Group, Los Angeles, California 90027 Judith Anne Stitt Department of Human Oncology, University of Wisconsin, Madison, Wisconsin 53792 Jeffrey F. Williamson Department of Radiation Oncology, Mallinckrodt Institute of Radiology, Saint Louis, Missouri 63110 ~Received 11 July 1997; accepted for publication 4 August 1997! Recommendations of the American Association of Physicists in Medicine ~AAPM! for the practice of brachytherapy physics are presented. These guidelines were prepared by a task group of the AAPM Radiation Therapy Committee and have been reviewed and approved by the AAPM Science Council. © 1997 American Institute of Physics.@S0094-2405~97!01410-7# Key words: brachytherapy, interstitial brachytherapy, intracavitary brachytherapy, remote afterloaders, quality assurance TABLE OF CONTENTS PART 1. INFORMATION FOR RADIATION ONCOLOGY ADMINISTRATION....... 1558 PART 2. A CODE OF PRACTICE FOR BRACHYTHERAPY PHYSICS.......... 1562 I. OVERVIEW................................ 1562 II. QUALITY ASSURANCE PROGRAM GOALS... 1563 A. Quality assurance program endpoints......... 1563 1. Safety of the patient, the public, and the institution............................ 1563 2. Positional accuracy..................... 1564 3. Temporal accuracy..................... 1564 4. Dose delivery accuracy................. 1565 B. Developing a quality assurance program...... 1565 1. Quality assurance of treatment delivery devices.............................. 1565 2. Procedure-specific quality assurance....... 1565 III. PHYSICAL QUANTITIES IN BRACHYTHERAPY........................ 1566 A. Brachytherapy source strength............... 1566 B. Source strength calibration.................. 1567 1. Conventional strength sources for LDR applications........................... 1567 2. High strength sources for HDR applications .................................... 1569 C. Single source dosimetry data................ 1570 1. ICWG formalism...................... 1570 2. Sievert integral model.................. 1570 3. Interstitial source data.................. 1570 4. Cesium intracavitary source data.......... 1571 5. HDR and PDR iridium-192 sources....... 1571 6. Intersource, heterogeneity, and applicator effects on dose........................ 1571 D. Source localization........................ 1572 IV. IMPLANT DESIGN AND EVALUATION...... 1573 A. Manual methods.......................... 1573 1. Manchester and Quimby systems......... 1573 2. Memorial nomographs.................. 1574 3. Paris system.......................... 1574 B. Computer methods of implant design......... 1574 1. Applicator-based planning............... 1575 2. Image-based planning................... 1575 C. Dose planning and evaluation............... 1576 1. Matched peripheral dose ~MPD!.......... 1576 2. Maximum continuous-contour dose for tumor bed implants..................... 1576 3. Maximum continuous contour dose for volume implants....................... 1576 4. Dose volume histograms................ 1577 5. Quality quantifiers..................... 1577 D. Dose specification and reporting............. 1578 1. ICRU recommendations for intracavitary brachytherapy......................... 1578 2. Recommendations for intracavitary brachytherapy......................... 1578 3. ICRU recommendations for interstitial brachytherapy......................... 1579 1557 1557 Med. Phys. 24 (10), October 1997 0094-2405/97/24(10)/1557/42/$10.00 © 1997 Am. Assoc. Phys. Med.

Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

  • Upload
    buikhue

  • View
    232

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

Code of practice for brachytherapy physics: Report of the AAPMRadiation Therapy Committee Task Group No. 56

Ravinder NathDepartment of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut 06510

Lowell L. AndersonDepartment of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York 10021

Jerome A. MeliDepartment of Radiology, St. Vincents Medical Center, Bridgeport, Connecticut 06606

Arthur J. OlchDepartment of Radiation Oncology, Southern California Permanente Medical Group, Los Angeles,California 90027

Judith Anne StittDepartment of Human Oncology, University of Wisconsin, Madison, Wisconsin 53792

Jeffrey F. WilliamsonDepartment of Radiation Oncology, Mallinckrodt Institute of Radiology, Saint Louis, Missouri 63110

~Received 11 July 1997; accepted for publication 4 August 1997!

Recommendations of the American Association of Physicists in Medicine~AAPM! for the practiceof brachytherapy physics are presented. These guidelines were prepared by a task group of theAAPM Radiation Therapy Committee and have been reviewed and approved by the AAPM ScienceCouncil. © 1997 American Institute of Physics.@S0094-2405~97!01410-7#

Key words: brachytherapy, interstitial brachytherapy, intracavitary brachytherapy, remoteafterloaders, quality assurance

TABLE OF CONTENTS

PART 1. INFORMATION FOR RADIATIONONCOLOGY ADMINISTRATION. . . . . . . 1558

PART 2. A CODE OF PRACTICE FORBRACHYTHERAPY PHYSICS. . . . . . . . . . 1562

I. OVERVIEW. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .1562II. QUALITY ASSURANCE PROGRAM GOALS... 1563

A. Quality assurance program endpoints. . . . . . . . . 15631. Safety of the patient, the public, and the

institution. . . . . . . . . . . . . . . . . . . . . . . . . . . .15632. Positional accuracy. . . . . . . . . . . . . . . . . . . . .15643. Temporal accuracy. . . . . . . . . . . . . . . . . . . . .15644. Dose delivery accuracy. . . . . . . . . . . . . . . . . 1565

B. Developing a quality assurance program. . . . . . 15651. Quality assurance of treatment delivery

devices. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .15652. Procedure-specific quality assurance. . . . . . . 1565

III. PHYSICAL QUANTITIES INBRACHYTHERAPY. . . . . . . . . . . . . . . . . . . . . . . .1566

A. Brachytherapy source strength. . . . . . . . . . . . . . . 1566B. Source strength calibration. . . . . . . . . . . . . . . . . . 1567

1. Conventional strength sources for LDRapplications. . . . . . . . . . . . . . . . . . . . . . . . . . .1567

2. High strength sources for HDR applications. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .1569

C. Single source dosimetry data. . . . . . . . . . . . . . . . 15701. ICWG formalism. . . . . . . . . . . . . . . . . . . . . .15702. Sievert integral model. . . . . . . . . . . . . . . . . . 1570

3. Interstitial source data. . . . . . . . . . . . . . . . . . 15704. Cesium intracavitary source data. . . . . . . . . . 15715. HDR and PDR iridium-192 sources. . . . . . . 15716. Intersource, heterogeneity, and applicator

effects on dose. . . . . . . . . . . . . . . . . . . . . . . .1571D. Source localization. . . . . . . . . . . . . . . . . . . . . . . .1572

IV. IMPLANT DESIGN AND EVALUATION . . . . . . 1573A. Manual methods. . . . . . . . . . . . . . . . . . . . . . . . . .1573

1. Manchester and Quimby systems. . . . . . . . . 15732. Memorial nomographs. . . . . . . . . . . . . . . . . . 15743. Paris system. . . . . . . . . . . . . . . . . . . . . . . . . .1574

B. Computer methods of implant design. . . . . . . . . 15741. Applicator-based planning. . . . . . . . . . . . . . . 15752. Image-based planning. . . . . . . . . . . . . . . . . . . 1575

C. Dose planning and evaluation. . . . . . . . . . . . . . . 15761. Matched peripheral dose~MPD!. . . . . . . . . . 15762. Maximum continuous-contour dose for

tumor bed implants. . . . . . . . . . . . . . . . . . . . .15763. Maximum continuous contour dose for

volume implants. . . . . . . . . . . . . . . . . . . . . . .15764. Dose volume histograms. . . . . . . . . . . . . . . . 15775. Quality quantifiers. . . . . . . . . . . . . . . . . . . . .1577

D. Dose specification and reporting. . . . . . . . . . . . . 15781. ICRU recommendations for intracavitary

brachytherapy. . . . . . . . . . . . . . . . . . . . . . . . .15782. Recommendations for intracavitary

brachytherapy. . . . . . . . . . . . . . . . . . . . . . . . .15783. ICRU recommendations for interstitial

brachytherapy. . . . . . . . . . . . . . . . . . . . . . . . .1579

1557 1557Med. Phys. 24 (10), October 1997 0094-2405/97/24(10)/1557/42/$10.00 © 1997 Am. Assoc. Phys. Med.

Page 2: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

4. ABS recommendations for interstitialbrachytherapy. . . . . . . . . . . . . . . . . . . . . . . . .1579

5. AAPM recommendations for interstitialbrachytherapy. . . . . . . . . . . . . . . . . . . . . . . . .1580

V. PERFORMING A BRACHYTHERAPYPROCEDURE. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .1580A. Initial planning. . . . . . . . . . . . . . . . . . . . . . . . . . .1580B. Treatment prescription. . . . . . . . . . . . . . . . . . . . .1580C. Ordering sources. . . . . . . . . . . . . . . . . . . . . . . . . .1581D. Receiving sources. . . . . . . . . . . . . . . . . . . . . . . . .1581E. Checking sources. . . . . . . . . . . . . . . . . . . . . . . . .1582F. Source and applicator preparation. . . . . . . . . . . . 1582

1. Iridium-192 seeds. . . . . . . . . . . . . . . . . . . . . .15822. Iodine-125 and palladium-103 seeds; other

permanently implanted seeds. . . . . . . . . . . . . 15823. Cesium-137 tube sources. . . . . . . . . . . . . . . . 1582

G. Loading applicators and sources. . . . . . . . . . . . . 15821. Iridium ribbons. . . . . . . . . . . . . . . . . . . . . . . .15832. Cesium tube sources. . . . . . . . . . . . . . . . . . . . 15833. Iodine-125 seeds and palladium-103; other

permanently implanted seeds. . . . . . . . . . . . . 15834. Remote afterloading. . . . . . . . . . . . . . . . . . . . 1583

H. Removal of sources, their security and returnto the vendor. .. . . . . . . . . . . . . . . . . . . . . . . . . . .15841. Permanent seed implants. . . . . . . . . . . . . . . . 15842. High dose rate remote after loaders. . . . . . . 1584

I. Source localization. . . . . . . . . . . . . . . . . . . . . . . .1584J. Treatment evaluation. . . . . . . . . . . . . . . . . . . . . .1584

1. Computer plan output. . . . . . . . . . . . . . . . . . . 15852. Special considerations for HDR

brachytherapy. . . . . . . . . . . . . . . . . . . . . . . . .1585K. Recording of physics data and other pertinent

information in patient chart. . . . . . . . . . . . . . . . . 1585VI. RECOMMENDED QUALITY ASSURANCE

PROGRAM FOR BRACHYTHERAPYEQUIPMENT. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .1585

A. Manual afterloading brachytherapy. . . . . . . . . . . 1586B. Remote afterloading brachytherapy devices. . . . 1587

1. Daily remote afterloader QA protocol. . . . . . 15892. Quarterly remote afterloader QA protocol. . 15913. Acceptance testing and annual remote

afterloader QA. . . . . . . . . . . . . . . . . . . . . . . .1592C. Quality assurance for treatment planning and

evaluation systems. . . . . . . . . . . . . . . . . . . . . . . .1592ACKNOWLEDGMENT . . . . . . . . . . . . . . . . . . . . . . . .1593APPENDIX A: DEFINITION OF A QUALIFIEDMEDICAL PHYSICIST . . . . . . . . . . . . . . . . . . . . . . . .1593APPENDIX B: BRACHYTHERAPY TEAMMEMBERS AND THE RESPONSIBILITIES OFTHE MEDICAL PHYSICIST . . . . . . . . . . . . . . . . . . . 1594APPENDIX C: INSTRUMENTATION NEEDEDFOR A BRACHYTHERAPY PHYSICSPROGRAM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .1594APPENDIX D: MAJOR REFERENCE TO NRCDOCUMENTS REGARDINGBRACHYTHERAPY . . . . . . . . . . . . . . . . . . . . . . . . . .1595

PART 1. INFORMATION FOR RADIATIONONCOLOGY ADMINISTRATION

The present document has been developed in a style similarto the 1994 AAPM code of practice for radiotherapyaccelerators,1 which was devoted to x-ray and electron beamradiotherapy machines. It is intended to cover similar issuesrelated to brachytherapy. Part I of this document is addressedto the radiation oncology administration, which may includea chief radiation oncologist, radiation oncology departmentadministrator, or a hospital/free-standing center administra-tor. In these guidelines, the AAPM recognizes the impor-tance of a team effort by administrators, radiation oncolo-gists, medical physicists, dosimetrists, radiation therapists,health physicists, and engineers in establishing an optimalbrachytherapy program.

Brachytherapy is the clinical use of small encapsulatedradioactive sources at a short distance from the target volumefor irradiation of malignant tumors or nonmalignant lesions.It plays an important role in the management of cancers ofseveral sites, including the brain, head and neck, uterine cer-vix, endometrium, and prostate. Recently, there is growinginterest in using brachytherapy for reducing restenosis aftertreatment for vascular diseases. Compared to conventionalexternal beam therapy, the physical advantages of brachy-therapy result from a superior localization of dose to thetumor volume. On the other hand, the dose gradients aroundan implant and dose heterogeneity within an implant aremuch higher than those in external beam radiotherapy. Un-like external beam fractionated radiotherapy, in low-doserate brachytherapy, radiation is continuously delivered overan extended period of time. There are two forms of brachy-therapy: intracavitary brachytherapy uses radioactive sourcesplaced in body cavities in close proximity to the tumor; andinterstitial brachytherapy uses radioactive seeds implanteddirectly into the tumor volume. Intracavitary brachytherapyis always temporary and usually takes from one to four days.On the other hand, interstitial brachytherapy can be tempo-rary or permanent. Also, several manual and remote-controlled afterloading techniques have been introduced toreduce radiation exposure to medical personnel. Remote af-terloaders, which have become very popular in the last tenyears or so, provide the ability to irradiate tumors at a varietyof dose rates from high-dose rate to conventional low-doserate in a continuous or pulsed sequence. High-dose rate af-terloaders provide brachytherapy as an outpatient procedurein many cases. Comprehensive radiation oncology servicesshould have access to a remote afterloader.

Over the past two decades, great strides have been madein the technology of diagnostic imaging as a basis for tumorlocalization, in the physics of radiation dosimetry, incomputer-assisted radiation treatment planning, and in thetechnology of external beam radiation machines and remoteafterloading brachytherapy. These technological develop-ments offer a wider spectrum of brachytherapy sources andtechnical capabilities, with new therapeutic possibilities.However, they pose new questions and problems to not onlythe radiation oncologist and the physicist, but also to the

1558 Nath et al. : Code of practice for brachytherapy physics 1558

Medical Physics, Vol. 24, No. 10, October 1997

Page 3: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

institution’s management team. Decision making in regard tonew brachytherapy facilities involves many individuals withdifferent expertise. It should start with the formulation of theradiation oncology needs of the institution based on the ex-pected patient population and include the development ofspecifications for all proposed equipment, housing and sup-port requirements, selection of the equipment itself, accep-tance testing, commissioning, quality assurance, mainte-nance, and finally initial and continual staff training.Compliance with state and federal regulations, as well asrecommendations from bodies such as the National Councilon Radiation Protection and Measurements~NCRP!, must beassured. In this document, where we differ on procedures orpractices currently mandated by regulatory agencies, a foot-note has been added to highlight the difference.

Brachytherapy treatment techniques are highly variablewith respect to their complexity, the extent to which they areindividualized to particular patients, and the degree to whichthey rely on prospective planning and dose calculations. Onone end of the spectrum are manually afterloading intracavi-tary procedures, utilizing relatively simple devices~a fixedpermanent radioactive source inventory and applicators!, andstraightforward manual calculations to define the loadingsand treatment times for individual patients. In contrast, manyrecently developed brachytherapy techniques heavily utilizeadvanced technology for target localization, for planning andoptimizing the proposed implant geometry, and for deliveryand verification of the treatment itself. For example, three-dimensional localization of the target volume by magneticresonance~MR!, computed tomography~CT!, ultrasound, orother imaging modalities is now standard of practice for im-plantation of tumors of the brain, prostate, and eye. The useof remote afterloading technology for both inpatient-andoutpatient-based brachytherapy is rapidly growing, as is theuse of image-guided applicator positioning technologies,e.g., stereotactically guided brain tumor implantation. Thishas been accompanied by rapid growth in the functionalityand complexity of treatment planning software. Currentlyavailable systems commonly support improvement of im-plant quality by optimization of individual dwell times, cor-relation of dose distributions with imaging studies, and con-trol of the remote afterloader during treatment delivery, inaddition to the classical function of computing and display-ing dose distributions.

Such technology-intensive treatment planning and deliv-ery techniques offer the prospects of improved clinical out-come~in terms of improved local control and reduced com-plications!, improved cost effectiveness, enhanced patientconvenience, and, in some cases, a more conservative, organ-preserving treatment alternative to more morbid and disfig-uring radical surgical approaches. However, such improve-ments come at the price of increased complexity, increasedrisk of serious treatment delivery errors and system malfunc-tions, and increased utilization of medical physicists, dosim-etrists, therapists, health physicists, and radiation oncolo-gists. Safe and effective use of any brachytherapy technique,however simple or complex, requires the involvement of aqualified medical physicist to~1! design and implement a

brachytherapy facility that meets the clinical needs of theinstitution, ~2! develop and implement treatment deliveryprocedures~for each clinical site and type of brachytherapyprocedure! that accurately realize the clinical intent of theradiation oncologist, protect the patient from treatment deliv-ery errors, maximize safety of the patient and staff, and,finally, minimize the legal and regulatory liability of the in-stitution, and~3! ensure the accuracy and safety of each in-dividual brachytherapy treatment through review of calcula-tions, monitoring treatment team compliance to establishedprocedures, and adapting procedures to meet the needs ofunusual patients.

Each of the major roles listed above will be reviewedbriefly in the following paragraphs. It is important for admin-istrators to understand that brachytherapy treatment deliveryis a team effort consisting of the medical physicist, alongwith appropriate support staff~dosimetrists, therapists, healthphysicists, and, in some cases, nurses! working in concertwith the radiation oncologist to accurately and safely deliverthe prescribed treatment. The physicist effectively serves asthe leader of the team with respect to planning and treatmentdelivery, determining which tasks and quality assurancechecks can be delegated to the team members, and whichcompleted tasks require physicist review. For relativelysimple manual afterloading implants, tasks such as sourcepreparation, loading, room posting, and patient surveys canbe assigned to support staff, and the direct role of the physi-cist limited to verification of treatment time and dose calcu-lations, periodic quality assurance, and periodic record au-dits. On the other extreme, high-dose rate brachytherapyprocedures and procedures requiring an implant to meetcomplex dosimetric specifications may require extensive in-volvement of the physicist in each case. The amount ofphysicist time and expertise required will depend on manyvariables, including~1! the sophistication of the technologyused in planning and delivering treatment,~2! the extent towhich implant dose distributions must be individualized toparticular patients and the complexity of the optimizationendpoints specified by the radiation oncologist,~3! the extentto which complex planning and delivery tasks have been‘‘proceduralized’’ so that they can safely be assigned to sup-port staff,~4! availability and sophistication of support staff,and ~5! availability of labor-saving technology, such ascomputer-assisted optimization, which can eliminate theneed for the time consuming and laborious manual optimi-zation.

‘‘Facility’’ is used here in a general sense to include allpermanent resources needed to implement the desiredbrachytherapy program including technical support person-nel, treatment planning and delivery equipment, and dedi-cated space such as high-dose rate brachytherapy procedurerooms and inpatient rooms needed for delivery of low-doserate brachytherapy. Facility design begins with the specifica-tion of sources, applicators, treatment delivery systems, soft-ware packages, and other needs. This involves close collabo-ration between the physicist and radiation oncologist toformulate the clinical needs of the program, the expectedcase load, and to identify specific pieces of equipment that

1559 Nath et al. : Code of practice for brachytherapy physics 1559

Medical Physics, Vol. 24, No. 10, October 1997

Page 4: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

meet this need. Specification and identification of appropri-ate spaces for treatment delivery, source storage, and prepa-ration is a task that requires close collaboration between thephysicist and appropriate representatives of the hospital ad-ministration. An important endpoint is protection of person-nel and visitors who occupy the spaces surrounding the treat-ment and source preparation facilities. A physicist is the onlyprofessional qualified to perform an analysis of the expectedworkload, and to design a system of structural or portableshielding to ensure that no staff or visitors receive exposuresin excess of limits proscribed by Federal and state regula-tions.

Another important function of the physicist is licensingthe proposed facility with the appropriate regulatory agency.In the U.S., medical use of reactor by-products, which in-cludes virtually all radionuclides used in clinical brachy-therapy, is very tightly and aggressively regulated at presentby the U.S. Nuclear Regulatory Commission~NRC!. Ap-proximately two-thirds of the states, called agreement states,have negotiated an agreement with the NRC in which day-to-day NRC enforcement and licensing activities are as-sumed by an appropriate state agency governed by stateregulations that parallel those of the NRC. In agreementstates, the hospital must license its brachytherapy activitieswith the appropriate state agency.

All brachytherapy programs must be conducted under theauthority of a license, i.e., a document that specifies the ra-dioactive materials that may be possessed, their alloweduses, and the authorized users~usually board certified radia-tion oncologists! who may treat patients in the facility. Thelicense is a contractual agreement between the hospital~notthe physicians or the medical physicist! and the regulatoryagency; thus the hospital administration is ultimately respon-sible for compliance with the terms of license. All licensesrequire that the institution have a radiation safety committee,consisting of representative users and a senior managementrepresentative, which monitors the use of radioactive mate-rials in the hospital. The AAPM recommends that a brachy-therapy physicist should be included as a member of theradiation safety committee. The hospital must have a radia-tion safety officer~RSO! who is responsible for operating theassociated radiation safety program, which involves manyactivities, i.e., monitoring all occupationally exposed personsto ensure that their exposures are as low as reasonablyachievable~ALARA !; properly receiving, surveying, andlogging in all radioactive sources; and implementing a qual-ity management program~QMP! that complies with theregulations. The RSO is often a physician or a medicalphysicist in smaller programs. In large institutions, the RSOis usually a full-time professional health physicist.

In a small hospital that has not previously been a memberof the regulated community, the physicist is the only on-siteprofessional with the expertise to write a license applicationand to work with the administration to develop a radiationsafety program and associated administrative structure. Inlarge institutions the addition or enhancement of a brachy-therapy program can often be handled by means of a licenseamendment or an additional specific license. In this case the

medical physicist can work with the existing RSO and radia-tion safety committee to develop the application. The licensewill specify many procedural details that can have significantimpact on day-to-day clinical operations, including the typeand frequency of quality assurance procedures, nursing andoperator training requirements, and even how often nursingpersonnel must check the implant for correct positioning.Whatever details the licensee agrees to in the licensing pro-cess become binding rules with the force of federal law be-hind them. By using an experienced physicist to draft thetechnical parts of the license application, the crippling effectsof an overly restrictive license agreement can often beavoided.

Hospital administrators are warned to take very seriouslythe need to comply ‘‘to the letter’’ with all regulations andlicense requirements. The NRC and many agreement statecounterparts have adopted a zero tolerance stance toward theregulated community. Inspectors will cite and fine the insti-tution ~and in some cases, individuals! for violating regula-tions and deviations from license commitments. Through itsQMP regulations, the NRC has enlarged its domain to in-clude quality and accuracy of patient treatments, as well aspersonnel protection. Certain types of treatment delivery er-rors, called ‘‘misadministrations,’’ must be reported to theNRC within 24 hours, regardless of whether there is poten-tial for harm to the patient. A misadministration will almostcertainly result in a special NRC inspection of your facility,notices of license and federal law violations, possible levyingof fines and other punishments, such as publicizing the treat-ment error in the press, and requiring senior administrators toattend enforcement conferences at the NRC regional head-quarters. Responding to misadministrations can consumehundreds of staff hours. Fortunately, the likelihood of such ascenario can be reduced to negligible levels by allocatingsufficient time to a qualified medical physicist for developingand maintaining an appropriate quality assurance~QA! pro-gram. The main goal of the QA program is to protect thewell-being of the patient and staff; however, an importantsecondary goal is to protect the legal interests of the institu-tion.

The final components in implementing a brachytherapytreatment facility are installation, acceptance testing, andcommissioning of all equipment. Again, the level of exper-tise and amount of time necessary for these activities de-pends on the complexity of the technology involved and theextent of the institution’s established brachytherapy facili-ties. Installation can range from overseeing the constructionof a dedicated HDR treatment suite, including structuralshielding and dedicated imaging capability, to simply receiv-ing a new set of manual afterloading sources. Acceptancetesting and commissioning involve subjecting the newly in-stalled equipment to exhaustive performance testing to deter-mine whether the vendor’s technical specifications and theinstitution’s clinical specifications are met, and to collectwhatever physical and dosimetric data are required for clini-cal implementation of the system. This includes verifying thecalibration and internal dimensions of sources, and verifying

1560 Nath et al. : Code of practice for brachytherapy physics 1560

Medical Physics, Vol. 24, No. 10, October 1997

Page 5: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

the positional and temporal accuracy of remote afterloadingequipment.

In general, the medical physicist is the only professionalat the hospital qualified to perform the activities described inthis section, including resource specification, facility plan-ning, license preparation, and commissioning/acceptancetesting of brachytherapy equipment, as well as that of com-puterized treatment planning systems. Although the input ofsupport personnel should be sought when appropriate, ingeneral few, if any, of these activities can be delegated tosupport staff.

Following installation and acceptance testing of brachy-therapy equipment, the physicist shifts to the task of devel-oping detailed procedures for planning and executing theproposed patient treatments. This involves sketching out theflow of the proposed treatment, including preoperative plan-ning, applicator insertion, implant imaging, and dose calcu-lation, and finally, source preparation and treatment. At eachpoint in the delivery process, the information required, andactions and decisions required, should be identified.

Vulnerable decision points in the delivery process, wherehuman error or device failure could cause errors in sourcepositioning, duration of treatment, or dose delivery, shouldbe identified and appropriate redundant checks designed~QAchecks!. The physicist works closely with the radiation on-cologist and support staff to make sure all important logisticproblems are covered and all actions and decisions requiringphysician intervention, feedback, or approval are identified.The proposed delivery process is carefully reviewed from theperspective of NRC/agreement state regulations and licenserequirements so that all information required to documentcompliance is captured.

The next step in procedure development is to assign rolesand responsibilities to the support staff~dosimetrist, thera-pist, source curator!, including those activities that requiredirect involvement or interaction of the physicist. Develop-ment of written procedures, including emergency proce-dures, procedures for treatment planning, optimization, anddose specification, and appropriate forms and checklists isthe next step. Forms are usually developed to rationalize thefollowing activities: source receipt, calibration, inventory,and disposal; prescription; patient survey and source re-moval; simulation and source localization; periodic QA oftreatment delivery equipment; manual verification of com-puter dose calculations, and auditing of records as requiredby the QMP program. QA checklists are very useful for de-fining the procedure flow, training new staff, and document-ing regulatory compliance. All members of the treatment de-livery team must be trained in their function and have theopportunity to develop the necessary technical skills, e.g.,remote afterloader programming, treatment planning systemoperation. For very complex procedures, dry run rehearsalsmay be needed, and the physicist will need to work closelywith all treatment team members during the first few treat-ments until training and finetuning of the procedure are com-pleted. Appropriate training of personnel in the correct ex-ecution of brachytherapy procedures and the correct

operation/application of brachytherapy equipment/applicators is essential.

Finally, before a new facility is put into clinical service, aperiodic QA protocol should be developed. The purpose ofthis program is to ensure that all devices required for treat-ment planning and delivery~sources, afterloaders, treatmentplanning systems, localization systems! continue to functionas assumed by the treatment delivery protocol. This protocolusually consists of a subset of commissioning tests which areto be performed at fixed intervals~annually, quarterly,monthly, or daily!. Oftentimes, daily QA tests are assignedto the support staff, while the more involved test sequences,performed at longer intervals, remain the physicist’s respon-sibility. Checklists and forms can greatly improve efficiencyand completeness of QA testing, as well as automaticallydocumenting compliance.

Development of well-documented procedures is a com-plex and time-consuming activity. However, the payback interms of error-free, consistent, and efficient treatment deliv-ery is large. Although development of procedures is prima-rily the responsibility of the physicist, close collaborationwith the radiation oncologist and other members of the treat-ment delivery team is critical. In general, little of this devel-opment activity can be delegated to support staff.

Good medical practices for brachytherapy have been de-scribed by medical and physics organizations including theAAPM through its task force reports,2,3 the American Col-lege of Radiology through its practice standards forbrachytherapy,4–6 and the American Brachytherapy Society.7

Such standards describe the level of service, staffing, clinicalcompetence, and treatment recommendations needed to pro-mote a safe, successful brachytherapy program. A significantcommitment of personnel and equipment resources by thehospital administration is necessary to establish a brachy-therapy program to support this clinical team.

The decision to provide brachytherapy services must beaccompanied by the concomitant decision to have the re-quired dosimetry and treatment planning equipment with theappropriate staff of qualified radiation oncology physicists.This code of practice recommends the radiation oncologyphysicist be certified in radiation oncology physics by eitherthe American Board of Radiology, the American Board ofMedical Physics, or the Canadian College of Medical Phys-ics ~Appendix A!. We recommend that radiation oncologyfacilities be staffed at levels not less than the guidelinesgiven in the ‘‘Blue Book’’ ~the report of the Inter-SocietyCouncil for Radiation Oncology!.8 In order to have a goodquality brachytherapy program, we recommend that facilitieshave a full-time qualified radiation oncology physicist. Ifonly a part-time consulting physicist is used, he or sheshould provide radiation oncology physics services of a qual-ity necessary for state-of-the-art brachytherapy treatment.

As noted above, the physicist’s level of involvement ineach patient procedure can vary significantly. Use of high-dose rate remote afterloading technology has increased thephysicist’s involvement. Current NRC regulations require aphysicist to attend each treatment to ensure rapid and expertresponse to any emergency situation. For low-dose rate re-

1561 Nath et al. : Code of practice for brachytherapy physics 1561

Medical Physics, Vol. 24, No. 10, October 1997

Page 6: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

mote afterloading, an on-call physicist should be available torespond to any technical or safety problem. The responsibili-ties of the medical physicist and other team members arelisted in Appendix B.

It is necessary that the qualified radiation oncology physi-cist have the appropriate equipment and test instrumentationneeded for source calibration, acquisition of dosimetry datafor the treatment planning computer, and the required peri-odic QA tests. A list of the necessary equipment is given inAppendix C.

In summary, the decision to provide a community withbrachytherapy facilities not only involves a decision to enlistthe services of a radiation oncologist, but also means enlist-ing the services of a qualified radiation oncology physicistand providing appropriate brachytherapy instrumentation. Inaddition, adequate support staff such as medical dosimetristsis essential for a safe and cost-effective operation of thephysics service. It is stressed that proper brachytherapy treat-ment is a team effort, and communication among team mem-bers encourages quality assurance. Due to the larger degreeof interdepartmental coordination needed, i.e., nursing, diag-nostic imaging, surgery, etc., a higher level of cooperationcompared to external beam radiotherapy must be developed.

A few comments on terminology are in order. There arethree levels of imperatives distinguished in this report:

~1! SHALL OR MUST:These terms are applied when theimperative is dictated by law.

~2! RECOMMEND: Phrases like ‘‘we recommend’’ and‘‘requires’’ are intended to convey that the AAPM con-siders the procedure referred to as important. If modifi-cation is considered, we recommend that it would occuronly after careful analysis demonstrates that qualitywould not be degraded. When a tolerance level or fre-quency of testing is given, it can be assumed to be arecommendation or law.

~3! SHOULD: There are many aspects of QA where toler-ance levels and frequencies cannot be given, and inwhich quality can be maintained via many different ap-proaches. In these instances, which apply to many as-pects of QA, modals like ‘‘should’’ are used. TheAAPM recognizes the complexity of the treatment plan-ning and treatment process, and the inadvisability andimpossibility of giving precise direction to QA in thisrespect. However, the AAPM considered it important tosuggest avenues for such quality assurance.

In Part II of this document we present a code of practicefor brachytherapy physics, which is based upon principlesfor good medical practice and management of risk.

PART II. A CODE OF PRACTICE FORBRACHYTHERAPY PHYSICS

I. OVERVIEW

Brachytherapy had its beginnings in Europe in the late1890’s with the discovery of radioactive radium-226. Theearly application of radium to skin lesions demonstrated the

effectiveness of this new treatment technique. Since thattime, many radionuclides have been developed offering awide range of half-lives and radiations. There is also a largevariety of clinical instrumentation available today for imple-menting numerous types of brachytherapy procedures.

Brachytherapy with radium, cesium, cobalt, or iridiumsources has traditionally been given at dose rates of 0.4–0.8Gy/h, a level referred to as low-dose rate~LDR!. Using theseradionuclides, the early pioneers of radiation therapy devel-oped highly successful treatment schedules for gynecologicmalignancies. In the past few decades, high-dose rate~HDR!brachytherapy has been developed as an alternative to theLDR brachytherapy. HDR refers to a dose rate in excess of0.5 Gy/min, a rate commonly administered with linear accel-erators for external beam therapy.

Intracavitary brachytherapy is the placement of radioac-tive sources in an applicator that has been positioned in abody cavity, i.e., the uterus, vagina, etc. Acceptable curerates using intracavitary brachytherapy are dependent on be-ing able to deliver significant radiation doses to tissues atconsiderable distances from the cavity surface, e.g., to thepelvic walls in gynecological brachytherapy. To meet thesegoals, applicator modifications, insertion techniques, dosespecification, and fractionation schemes have been highlydeveloped for LDR and HDR intracavitary brachytherapysystems.

Intraluminal brachytherapy is the temporary placement ofa radioactive source or sources in a linear arrangement insidethe lumen. It is often used for tumors that obstruct the open-ing of a pulmonary bronchus, biliary duct, esophagus, etc.Catheters placed by endoscopy are afterloaded with radioac-tive sources to deliver a dose that can relieve the obstruction.

Interstitial brachytherapy is temporary or permanent im-plantation of radioactive seeds or needles directly in a tumorvolume. It is particularly suited for prostate, gynecologic,and locally recurrent cancers. Therapy is accomplished usingLDR or HDR techniques of manual or remote afterloading.In an afterloading technique, first hollow catheters or guidetubes are inserted in the target volume and they are loadedwith radioactive sources afterwards. Computerized dose cal-culation prior to interstitial brachytherapy is often necessaryfor delivering a homogeneous tumor dose and avoiding hotspots in the tumor and surrounding normal tissues.

This part of the code of practice presents the AAPM rec-ommendations on the physical aspects of all types of brachy-therapy, LDR, or HDR. After describing the goals of a qual-ity assurance program in Sec. II, the physical quantities usedin brachytherapy are described in Sec. III. The specificationof the strength of brachytherapy sources in terms of airkerma strength is recommended. Single source reference dataand a formalism for dose calculations for interstitial brachy-therapy are presented. Also described are the techniques forsource localization in the patient.

In Sec. IV, implant design and evaluation are described.Traditional dose specification systems such as the Paris,Manchester, and Stockholm systems of brachytherapy aredescribed first. Both image-based and applicator-based com-puterized planning systems are described next. In the later

1562 Nath et al. : Code of practice for brachytherapy physics 1562

Medical Physics, Vol. 24, No. 10, October 1997

Page 7: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

parts of Sec. IV, various methods of dose planning, evalua-tion, specification, and reporting are described.

Section V deals with the implementation of a brachy-therapy implantation. Initial planning, treatment prescription,ordering sources, receiving sources, checking sources, sourceand applicator preparation, loading applicators, removal ofsources, and return of sources to vendor are some of theissues discussed in this section. Finally, in Sec. VI, a qualityassurance program for brachytherapy is recommended.

This task group does not address the issues dealing withthe design and commissioning of brachytherapy facilities.This topic will be reviewed further by the radiation therapycommittee in a future report.

Much of the text in this report is descriptive in nature. Itreflects current of state-of-the-art in clinical brachytherapywith an extensive bibliography. Where appropriate, theAAPM makes specific recommendations regarding goodmedical practice of brachytherapy. Part of the objective ofthis report is educational and the other is to recommend astandard of practice for brachytherapy.

II. QUALITY ASSURANCE PROGRAM GOALS

The goal of the brachytherapy quality assurance~QA!program is to maximize the likelihood that each individualtreatment is administered consistently, that it accurately re-alizes the radiation oncologist’s clinical intent, and that it isexecuted with regard to safety of the patient and others whomay be exposed to radiation during the course of treatment.The QA program consists of a set of mandated redundantperformance checks, physical measurements, documentationstandards, training and experience standards, and guidelinesfor the development of treatment procedures that are de-signed to minimize the frequency of human errors, miscom-munication, misunderstandings, and equipment malfunc-tions. With respect to treatment delivery, accurate treatmentmeans that the intended sources are delivered to their in-tended positions within the correct applicator, remain therefor the correct length of time, and accurately deliver the ab-sorbed dose required to realize the radiation oncologist’swritten prescription. This of course presupposes that the im-plant design and evaluation results in a spatial–temporal dis-tribution of radioactivity consistent with the goals of treat-ment and with the applicator arrangement inserted into thepatient. A comprehensive QA program addresses each of thethree basic processes:

~1! Applicator insertion process.Applicator selection andplacement in the patient is under control of the radiationoncologist and referring physician: Its success depends onthe experience and surgical skill of these physicians. PhysicsQA duties include documentation of the applicator systeminserted, its correct operation, and correct correlation withtarget volume.

~2! Implant design and evaluation process.This processbegins with selection of applicator type and implant design.Following surgical realization of the implant, it continueswith formulation of the prescription, radiographic examina-tion of the implant, definition of the target volume, and

computer-assisted dose calculation and optimization. Theend result of this process is identification of the desiredspatial–temporal distribution of radioactivity needed to ful-fill the prescription. For manual brachytherapy, this includesapplicator loadings and durations. For remotely afterloadedbrachytherapy, the end result of treatment planning includesprogramming parameters for afterloading, i.e., the correctdwell positions and dwell times for each catheter. Correctparameters mean those required to accurately deliver the pre-scribed dose distribution, including any volume or dose con-straints on normal tissue irradiated. A QA program must en-sure, in general and for each treatment, that the treatmentplanning program functions accurately, that the system forinferring dwell locations from simulation radiographs per-forms accurately, that the target volume rendered on thesefilms is consistent with all known tumor localization data,and that optimization endpoints used by the treatment plan-ning program are appropriate.

~3! Treatment delivery process.For remote afterloading,the delivery process includes entry of the programming pa-rameters into the remote afterloader, connection of the pa-tient to the device, and delivery of treatment. The qualityassurance program must contain procedures for validatingthe entered data, responding to unexpected machine mal-functions and emergencies, and documenting the deliveredtreatment. For manual brachytherapy, treatment delivery in-cludes selection, preparation, and insertion of the sources aswell as removal of the sources at the designated time. For alltreatments, the delivery process includes procedures neces-sary for patient and staff safety throughout the process.

A. Quality assurance program endpoints

The variability in brachytherapy device features and clini-cal practice standards~see Part I! precludes development of afixed QA protocol. Therefore, from basic principles, eachphysicist must develop a program specifically suited to his orher individual clinical environment. Indeed, one of the chal-lenges of clinical brachytherapy physics is to identify therelevant quantitative endpoints and the accuracy with whichthey must be realized to carry out the radiation oncologist’sclinical intent in a practical and reasonable fashion. System-atic development of a QA program that encompasses bothdevice function and human factors requires that the clinicalgoals of the treatment program be identified, translated intophysical endpoints, and assigned tolerances for acceptableperformance under realistic practical situations. For anyclinical brachytherapy application, these endpoints fall intofour broad categories:

1. Safety of the patient, the public, and theinstitution

This QA endpoint addresses all populations whose wellbeing is potentially threatened by the brachytherapy pro-gram. Safety of the public and involved health care person-nel includes control of radiation exposure to staff and mem-bers of the public, adequacy of the facility shielding barriers,and procedures to maintain control of all radiation sources.Promotion of patient safety entails prevention of catastrophic

1563 Nath et al. : Code of practice for brachytherapy physics 1563

Medical Physics, Vol. 24, No. 10, October 1997

Page 8: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

treatment delivery errors, or other conditions that threatenthe well being of the patient, as a result of device malfunc-tion or human errors in the design, evaluation, and executionof the brachytherapy procedure. For remote afterloaders, pa-tient safety QA includes verifying correct function of rel-evant error recognition features, interlocks, treatment statusindicators, and emergency response systems. A safety QAcheck has the general form, ‘‘Does specified hazard X~e.g.,the source been transported to the wrong location! exist? If Xexists, then perform emergency response Y~e.g., halt thetreatment, retract the sources, and summon expert help! orelse continue the treatment.’’ Specification of the expectedemergency responses of automated treatment devices, or de-sired emergency procedures when prescribed human re-sponses are required, is an important element of the program.Protection of institutional safety involves minimizing condi-tions that create potential legal or regulatory liability, eventhough they pose no threat to patient care or staff well being.For example, making inaccurate entries into a patient’s treat-ment record, even when the information in question is clini-cally irrelevant to the particular patient, may increase theinstitution’s legal liability by calling the credibility of therecord keeping process into question in event of a futurelawsuit. Certain Nuclear Regulatory Commission~NRC!regulations~e.g., quarterly facility surveys for192Ir HDRunits! add little to the quality of patient care or staff safety,but must be completed and documented properly to avoidregulatory enforcement actions. By maintaining expertise inregulatory detail and documenting compliance to all regula-tions, the physicist makes an important contribution to insti-tutional safety.

Safety QA procedures and endpoints have been heavilyinfluenced by NRC~or agreement state counterpart! regula-tions. The governing guidelines are spelled out in great detailby the Nuclear Regulatory Commission in Title 10, Parts 20and 35 of the Code of Federal Regulations~10 CFR Part 20and 10 CFR Part 35!. Part 20 outlines area and personnelexposure limits as well as requirements pertaining to label-ing, storage, shipping, and handling of sealed sources. Part35 outlines the responsibilities of the Institutional RadiationSafety Committee and Radiation Safety Officer as well as theminimum credentials physicians must possess in order to beauthorized users, i.e., to be allowed to prescribe brachy-therapy to human patients. Part 35 contains many highly de-tailed regulations addressing, e.g., source inventory andcheck-out procedures, frequency and type of exposure sur-veys required, frequency and type of treatment record auditsrequired, and the entries required for brachytherapy prescrip-tions ~or ‘‘written directives’’ in NRC jargon!. Appendix Dlists NRC documents outlining essential regulatory, licens-ing, and compliance standards for brachytherapy.

With the rapid growth of regulatory initiatives in brachy-therapy and more aggressive intrusion into clinical practice,a temptation to confuse adequate QA with regulatory com-pliance is understandable. This is a serious error. RegulatoryQA and safety mandates, in general, are neither sufficient nornecessary conditions of accurate and safe brachytherapytreatment. For example, while NRC strictly regulates the

calibration procedures for survey meters, there are no re-quirements regarding accuracy or traceability for LDRbrachytherapy source calibrations. A major task of thebrachytherapy physicist is development and maintenance ofa QA program that ensures good medical practice standards:Regulatory compliance is only one component of this largerand more general mandate.

2. Positional accuracy

Verification of positional accuracy requires that one con-firms that the intended sequence of active sources or dwellpositions is delivered to the correct position in the correctapplicator. ‘‘Correct position’’ refers specifically to the po-sitions defined directly or indirectly by the attending radia-tion oncologist. Often, the target source locations are identi-fied relative to radiographic images of dummy seeds orradiographic markers which are inserted into the applicatorof interest prior to simulation. For surface-dose or gyneco-logical intracavitary applicators, the correct position may bedefined as the expected position relative to the applicatorsurfaces. Positional accuracy assessment reduces to verifyingthe protocol~hereafter called simulation source localizationprocedure! for calculating the source loading instructionsused to position the actual source at a desired location in thecatheter defined by the radiographic marker seeds. When re-mote afterloading devices are used, these instructions aremachine programming parameters~length, position, channelnumber!. In most clinical applications of afterloaders, a po-sitional accuracy of62 mm relative to the applicator system~not anatomical landmarks in the patient! is reasonable.@Note that for remote afterloaders, the NRC insists on a po-sitional accuracy criterion of61 mm ~policy and guidancedirective FC 86-4!. This more rigid standard is not realizablein a clinically meaningful sense for many applicator-sourcecombinations.#

3. Temporal accuracy

A treatment system achieves temporal accuracy if eachsource sequence or single source dwell position remains atits intended position for the length of time specified by thetreatment program. For manually afterloaded temporary im-plants, the goal is to develop a procedure to ensure that theradioactive sources are removed upon completion of dosedelivery. Remotely afterloaded brachytherapy places treat-ment duration under control of an electronic timer. Tests ofabsolute timer accuracy are required whenever source cali-bration is based on an external time standard, whereas rela-tive tests suffice when the machine timer is used both tocontrol treatment delivery duration and to integrate chargemeasurements during source-strength calibration. An accu-racy criterion of 62% seems easily achievable, both bymanual afterloading techniques and commercially availableremote afterloading systems. In addition, the influence oftransit dose on dose delivery accuracy must be evaluated andcorrected for, if necessary. Transit dose is the additional dosedelivered while the source is in motion.

1564 Nath et al. : Code of practice for brachytherapy physics 1564

Medical Physics, Vol. 24, No. 10, October 1997

Page 9: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

4. Dose delivery accuracy

Even with completely error-free delivery of the spatial–temporal distribution of radioactivity, i.e., accurate realiza-tion of source positioning and treatment duration, many othervariables must be controlled in order to assure accurate de-livery of absorbed dose in tissue. It is useful to subdividedose delivery accuracy into physical and clinical aspects.Physically accurate dose delivery is achieved if the predicteddose and actual dose absorbed by the medium are equal atreference points specified without positional error relative tothe applicator. Physical dose delivery accuracy neglects thedifficult problem of defining dose calculation points relativeto patient anatomy. Accurate calibration of the source interms of a well-defined physical quantity, preferably airkerma strength, is among the most important physical dosedelivery parameters. Other factors include selection of accu-rate dosimetric data for calculating the single source dosedistribution and the influence of applicator attenuation andshielding corrections on the dose distribution. It is difficult toassign a meaningful tolerance to this endpoint as no practicaland validated dose measurement technology is available tothe hospital physicist. However, a source calibration accu-racy of 63% relative to existing air kerma strength stan-dards seems reasonable. Based on recent low-dose rate do-simetry experience, physical dose delivery accuracy on theorder of 5–10% is achievable at distances of 1–5 cm frommost common LDR sources. Finally, relative to the inputdata supplied and the algorithm assumed, the computer-assisted dose calculations should have a numerical accuracyof at least62%.

Clinical dose delivery accuracy includes a large array ofoften difficult-to-solve problems. Relatively straightforwardissues include the accuracy with which the dosimetrist andtreatment planning computer reconstruct the relative three-dimensional geometry of the implant. In intracavitarybrachytherapy, consistent, if not accurate, localization ofbladder and rectal reference points is often important. Fi-nally, if dwell weights are optimized to achieve dose unifor-mity or adequate coverage of a specified target volume, care-ful attention to optimization endpoints, prescription criteria,and quality of the resultant implant is required. The moredifficult problems include identification of the target volumeand critical organ margins relative to the implanted applica-tors and controlling or compensating for patient motion.Clearly, if uncertainties in the clinical procedure are large,then the accuracy of physical dose delivery is less important.The tolerance level, therefore, should be determined by eachuser based on practical considerations.

B. Developing a quality assurance program

Most errors in brachytherapy are the result of human er-rors, miscommunications, or misunderstanding of equipmentoperation rather than failure of the treatment delivery andplanning devices to perform properly. Brachytherapy treat-ment planning and delivery are complex human activitiesinvolving cooperation of physician, physicist, technologist,nurse, and dosimetrist, all of whom must execute their func-

tions correctly and must unambiguously receive and transfercritical information from one another. A team approach isrecommended. All team members should be encouraged todouble check each other and identify problems without fearof retribution. Error identification should be praised and re-warded as a sign of good QA. QA program developmentmost focus on~1! correct function and physical characteris-tics of treatment planning and delivery devices~includingsources, afterloaders, dose calculation tools, and test instru-ments! and ~2! the correct execution of each brachytherapyprocedure, i.e., procedure specific QA.

1. Quality assurance of treatment delivery devices

Device QA, covered in Sec. VI of this report, includesinitial acceptance testing and commissioning of devices. Itspurpose is not only to determine whether each system func-tions as specified, but to identify its operational characteris-tics, to give the physicist an opportunity to become an expertuser, and to develop procedures for using the device to treatpatients. A program of QA checks to be repeated at pre-scribed intervals must be put into place. The goal of periodicdevice QA is to confirm that the important operating charac-teristics of the device remain unchanged through time. Otheraspects of system-wide QA include developing training ma-terials and curricula for nurses, dosimetrists, and therapistsinvolved in treatment delivery, as well as verifying throughanalysis of attendance records, that training remains current.

2. Procedure-specific quality assurance

A procedure-specific QA protocol is a set of specifiedactions selected to ensure that each important step leading todelivery of a brachytherapy procedure is correctly carriedout. Often the specified actions are redundant tests andchecks designed to confirm correct execution and activity.More commonly, QA guidelines are rules or procedures thatdefine the procedure chronology or restrict the range of ac-tions that are possible at any point, thereby limiting the typesof errors that can be made. For example, insisting that onlynurses with radiation safety/radioactive procedures trainingcan care for brachytherapy patients or that a medical physi-cist must perform certain types of calculations reduces thelikelihood of error by requiring that expert personnel beavailable at specified points in the treatment delivery processto ensure that key actions are executed with a high level ofconfidence. Similarly, requiring that the radiation oncologistcomplete the written prescription before initiating the treat-ment evaluation process minimizes the likelihood that iso-dose calculation will be based on incorrect source loadingsor prescribed doses. QA program development involves notonly development of redundant checks, but carefully design-ing the flow of the procedure itself to minimize the likeli-hood of serious errors. Because practice standards and com-plexity of treatment planning and design are so variable,procedure-specific QA is highly individualized not only tothe institution but to each type of procedure. Commonly ac-cepted procedure-specific QA elements are reviewed in Secs.IV, V, and VI.

1565 Nath et al. : Code of practice for brachytherapy physics 1565

Medical Physics, Vol. 24, No. 10, October 1997

Page 10: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

To develop an effective procedure-specific QA program,the AAPM recommends the following step-by-step proce-dure:

~1! Define the anticipated or actual flow of the procedure,including all major steps~e.g., implant design, applicatorinsertion, implant imaging, implant evaluation, etc.!. Ateach step, identify the involved team members~physi-cian, therapist, etc.!, the critical activities to be per-formed and the information that must be captured.

~2! The AAPM strongly recommends use of carefully de-signed forms for capturing and documenting all criticalinformation, including the implant drawing, applicatorsutilized, catheter numbering system, target localizationdata, and written prescription. Easy-to-use documenta-tion will ensure accurate communication among brachy-therapy team members~e.g., from the physician to thephysicist and treatment planner!, among the variousphysical locations involved~e.g., operating room to im-aging suite!. In addition, such written documentation~2–5 forms, depending on procedure complexity!, formsthe basis of the patient’s permanent treatment record.

~3! Identify vulnerable points in the treatment delivery pro-cess, where mistakes, misjudgments, or inaccurate trans-mission of data can jeopardize the outcome of the pro-cedure. A redundant check should be designed,specifying who is to perform the check and what actionsare to be taken if the test result deviates from the ex-pected outcome. Both severity and likelihood of the tar-get error should be taken into account in deciding how todistribute available QA resources. Low probability cata-strophic scenarios~e.g., failure of HDR source to retract!should not be emphasized to the exclusion of more com-mon but less severe human errors~e.g., misidentificationof source strength, erroneous estimation of source posi-tioning parameters from simulation films!.

~4! Develop a written procedure, outlining the brachy-therapy procedure chronology, team member functions,QA checks, and associated documentation. Each brachy-therapy practice should develop written procedures andpatient-specific documentation for each major type ofprocedure, as described above. In addition, each institu-tion should develop a mechanism, formal or informal,for confirming compliance with the written QA program.One approach is to formalize the execution of the treat-ment delivery process by means of QA check-off forms.Developing such forms forces one to systematically con-ceptualize the treatment delivery process. In addition todocumenting compliance with QA requirements, check-off forms are useful for training new team members, andfor guiding the flow of infrequently performed proce-dures. However, the AAPM recognizes that formal docu-mentation of all QA checks constitutes a significant bur-den and is not the only approach to confirming QA. Fora highly experienced treatment delivery team, which fre-quently performs brachytherapy procedures, QA check-lists may not be as useful and mandatory. In such cases,the alternative approach should be clearly outlined.

~5! The AAPM further recommends that the brachytherapyQA program should be integrated into the overall depart-mental QA program~QA system! as defined by AAPMTask Group No. 40.3 This assignment gives the depart-mental QA committee~QAC! responsibility for monitor-ing the performance of the brachytherapy QA programso that any shortcomings can be identified and corrected.The QAC can monitor brachytherapy QA system perfor-mance by independently auditing a sample of patientrecords and QA checklists to confirm that written QAprocedures are being followed. QAC review of anybrachytherapy treatment delivery errors is another feed-back mechanism by which deficiencies in the QA pro-gram or its implementation can be identified. Anotherhighly desirable enhancement of the basic QA programis to institute a formal continuous quality improvement~CQI! process. Cost effective utilization of QA re-sources, as well as minimization of treatment deliveryerrors, is another QAC function. After finding that a par-ticular QA test reveals an acceptably low incidence er-rors, the QAC might recommend dropping the test orincreasing the action level.@The QA system recom-mended here is only superficially similar to the NRCquality management program~QMP! as described in 10CFR 35.32. The QMP is a highly prescriptive rule defin-ing precise endpoints, checks, auditing, and review pro-cedures. The NRC’s QMP goal is very limited: ‘‘...toprovide high confidence that byproduct material ... willbe administered as directed by the authorized user.’’ Incontrast, the QA system as defined and endorsed by theAAPM in this document allows institutions wide latitudein defining QA endpoints, documentation standards, QAtest methodologies, test outcome action levels, auditing,and other oversight mechanisms.#

III. PHYSICAL QUANTITIES IN BRACHYTHERAPY

A. Brachytherapy source strength

Source-strength designation has gone through severalchanges over the years.9 The earliest quantity~mass of ra-dium! was commonly referred to by the unit, milligram ra-dium. It was later generalized to milligram radium equivalentfor other radionuclides, and this quantity~equivalent mass ofRa! is still in use. Milligram radium equivalent~mgRaEq!means that the source so designated produces an exposurerate in free space at a large distance on its transverse axis,equal to that for the same mass of radium encased in a cap-sule of 0.5-mm Pt wall thickness. A ‘‘large’’ distance, in thiscontext, means large enough that the inverse square law isobeyed. Although this designation could be used for any ra-dionuclide, it is meaningful only for those emitting high-energy photons, such as137Cs, 192Ir, etc. Equivalent strengthsources of these radionuclides will yield nearly equal doserates in tissue along their transverse axes. However, for low-energy photon emitters such as125I, the attenuating effect oftissue is much greater such that an in-air mgRa equivalencydoes not translate to an equivalency of dose in tissue.

1566 Nath et al. : Code of practice for brachytherapy physics 1566

Medical Physics, Vol. 24, No. 10, October 1997

Page 11: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

Most treatment planning systems allow source strengthsto be specified as mgRa equivalent or in terms of the moregeneric quantity, activity. The latter is the number of disin-tegrations per second and is applicable to radionuclides ofany energy. However, this quantity is not widely used inbrachytherapy. For sealed sources, especially those of lowenergy, the encapsulation reduces the air kerma and doserates below those which would be produced by the baresource. Thus the strength is generally given as apparent ac-tivity, which is less than the encapsulated activity. Apparentactivity is the activity of a hypothetical point source of thesame radionuclide which would produce the same air kermarate, at the same large distance, as that measured on thetransverse axis of a sealed source. The design of the sourcecapsule also influences the dose distribution around thesource. It is quite possible for two sources of the same ra-dionuclide and same apparent activity to have different dosedistributions.

The emphasis today is to specify source strength by aNIST-traceable quantity, which is related to air kerma rate atsome distance in air. Therefore, the recommended sourcespecification is air kerma strengthSK given by

SK5Kl 2, ~1!

wherel is the reference distance at which the air kerma ratein free space,K, is specified. The unit of air kerma strengthis mGy h21 m2, which is numerically equivalent tocGy h21 cm2. This unit has been denoted by the symbol U.

A National Institute of Standards and Technology~NIST!calibration of a brachytherapy source is simply an in-air airkerma rate (K) measurement at a large distance (r 0) in freespace on the transverse axis. It does not specify activity,apparent or otherwise. However, it has been customary forsource vendors to convert the NIST-measured air kerma rateto apparent activity (A) usingA5K(r 0)r 0

2/GK , whereGK isthe air kerma rate constant. As a first step in a dose calcula-tion, the user might multiply the stated activity by aGK valueto obtain an air kerma rate. ThisGK must be the same as thatused by the vendor in order to restore the original NISTcalibration, even if that value has no basis in reality. In ef-fect, GK is a dummy parameter.

The American Association of Physicists in Medicine10

and the American Brachytherapy Society11 recommend thatair kerma strength be used at all levels in the brachytherapytreatment delivery process including ordering of sources,dose computation, treatment planning, treatment prescrip-tion, and implant documentation. Specifically, air kermastrength should be used to quantify source strength as fol-lows:

~1! All NIST, accredited dosimetry and calibration labora-tory ~ADCL!, and vendor certificates should use airkerma strength to describe strength of brachytherapysources. Calibration factors for institutional calibrationtransfer instruments such as re-entrant chambers shouldbe described in terms of air kerma strength.

~2! Input of data into computer-assisted treatment planningsystems, as well as printed output documentation, should

utilize air kerma strength. All treatment planning soft-ware vendors are urged to modify their dose calculationalgorithms and user interfaces so that all displayed andprinted references to source strength clearly and unam-biguously describe this quantity and its units. All manualdosimetry aids, such as nomograms, surface dose tables,and planning algorithms, should be normalized in termsof air kerma strength.

~3! All published dose distribution data for brachytherapysources should be normalized in terms of air kermastrength.

~4! Source loadings for individual patient implants should beprescribed and documented in terms of air kermastrength. All written brachytherapy treatment prescrip-tions requiring explicit reference to source strengthshould be stated in terms of air kerma strength. Pub-lished clinical studies in brachytherapy should utilizethis quantity to report source strength and applicatorloadings.

All members of the association are urged to work withintheir institutions to introduce and encourage use of air kermastrength in all clinical discussions, case presentations, andteaching conferences that involve discussion of brachy-therapy source strength.

It should be noted that two different source designs of thesame isotope and same air kerma strength can, particularlyfor low energies, produce different dose rates in tissue atequal distances along their transverse axes. For example,with 125I sources, the dose rate at 1 cm along the transverseaxis of model 6702 is approximately 8.5% greater than formodel 6711, even though both are identically encased. Thedifference is attributed to fluorescent x-rays from the silverwire in the model 6711. Thus the Interstitial CollaborativeWorking Group ~ICWG! recommended that does calcula-tions be based on the product of dose rate constant and thesource strength, defined to be the dose rate in medium perunit air kerma strength at a distance of 1 cm along the trans-verse axis.12 This quantity should be obtained from in-phantom measurements or calculations for each source de-sign. So far, this has been accomplished for the interstitialsources192Ir, 103Pd, and the two models of125I, resulting inrecommended dose rate constants for each.13 This dosimetryprotocol is further described in Sec. III C 1.

B. Source-strength calibration

1. Conventional strength sources for LDRapplications

As a result of the decreasing use of radium sources, NISTno longer maintains a standard for radium. At present, NISTprovides calibrations for most137Cs sources, several styles of192Ir, and125I sources. Air kerma strength standards for137Csand192Ir were established from exposure measurements at alarge distance using spherical graphite ionization chambersof known volume.14,15 In 1985, NIST established an airkerma strength standard for125I using a free-air ionizationchamber.16 However, it was subsequently realized that themeasured exposure included contributions from very low-

1567 Nath et al. : Code of practice for brachytherapy physics 1567

Medical Physics, Vol. 24, No. 10, October 1997

Page 12: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

energy fluorescent x rays originating in the titaniumcapsule.17,18 Since these nonpenetrating x rays do not con-tribute to the dose in tissue they should be excluded from theair kerma measurement. NIST is currently in the process ofrevising the calibration of125I to take this effect into account.

Standards for125I and 192Ir brachytherapy sources aretransferred by NIST to other sources through the use of alarge volume spherical well-type ionization chamber. A cali-brated source of a particular radionuclide is placed in thewell chamber in a fixed geometry. This serves to calibratethe chamber. The standard is transferred to other sources bymeasuring them in the well chamber for the same geometry.The well chamber has a 4p geometry, while the sourcespecification is for a point on the transverse axis. The cham-ber is therefore sensitive to the anisotropy of the air kermadistribution around the source. Thus calibration can be trans-ferred only to sources having essentially the same design asthose used in the initial free-air measurements. For137Cssources the transfer of calibration is performed using a largevolume chamber at a distance.

The NIST SK standard is transferred to the ADCLs in asimilar manner. A source of the proper design is sent toNIST for calibration and used to establish the standard forthe ADCL well-type ionization chamber. The NIST standardor the ADCLs standards are considered as national standards.A customer’s source is calibrated at an ADCL or NIST byplacing it in the well chamber. Alternately, a customer’s wellchamber is calibrated at an ADCL or NIST against a nationalstandard at an ADCL or NIST.

The AAPM Task Group No. 40 had recommended trace-ability of brachytherapy sources3 in 1994. The AAPM up-dates these recommendations here. All sources for whichNIST provides a calibration should have calibrations trace-able to NIST in one of the following ways:

~1! Direct traceabilityis established when either a source ora transfer instrument~e.g., well chamber! is calibratedagainst a national standard at an ADCL or at NIST itself.

~2! Secondary traceabilityis established when the source iscalibrated by comparison with the same radionuclide anddesign that has a directly traceable calibration or by atransfer instrument that bears a directly traceable calibra-tion.

~3! Secondary traceability by statistical inferenceis estab-lished when a source is one of a group of sources ofwhich a suitable random sample has direct or secondarytraceability.

For brachytherapy sources that do not have a nationalstandard yet users should develop a constancy check cali-brated against the vendor’s standard and use this constancycheck to verify the source strength. Another option is to de-velop one’s own secondary standard along the lines sug-gested by Goetschet al.,19 Das et al.,20 and Verhaegenet al.21

Well-type ionization chambers make it easy to establishsource calibrations with one of these traceabilities. Thus, forsources for which NIST provides calibration it is no longernecessary and should no longer be a practice to rely on

source strengths quoted by the manufacturer. At least onelong-lived source,226Ra or137Cs, should be kept in inventoryfor a quality control check on the well chamber’s responseprior to each use. A record maintained of the chamber’s re-sponse to evaluate its precision and long term stability.Variations of less than 1% around a mean should be achiev-able. For air-communicating chambers, it is important tomake temperature-pressure corrections, and for137Cs, to cor-rect the chamber reading for source decay.

Ideally, every radioactive source that is to be implanted ina patient should be calibrated. In practice however, limita-tions of time, personnel exposure, or other physical con-straints preclude this level of thoroughness. We recommendthat all long half-life sources be calibrated. Traceability bystatistical inference may be appropriate for short half-lifesources, depending upon the number of ribbons or seeds inthe designated strength groupings under consideration. If thegrouping contains only a few seeds we recommend the cali-bration of all seeds. For groupings with a large number ofloose seeds, we recommend that a random sample containingat least 10% of the seeds be calibrated; for a large number ofseeds in ribbons, a minimum of 10% or 2 ribbons~whicheveris larger! should be calibrated. For sources purchased in asterile configuration, we recommend purchasing and calibrat-ing a single~nonsterile! seed for each designated-strengthgrouping.

Brachytherapy sources are assigned a calibration by themanufacturer. Every institution practicing brachytherapyshall have a system for measuring source strength with sec-ondary traceability for all source types used in its practice.Prior to using newly received sources for treatment, thevendor-supplied~with the exceptions noted in the precedingparagraph! calibrations must be verified as per Task GroupNo. 40 recommendations.3 The institution should comparethe manufacturer’s stated value with the institution’s stan-dard. If the two are within acceptable limits~see Table I!,either the manufacturer’s or institution’s value may be used.We recommend that if the institution’s verification of sourcestrength disagrees with the manufacturer’s data by more than3%, the source of the disagreement should be investigated.We further recommend that an unresolved disparity exceed-ing 5% should be reported to the manufacturer. It is alwaysadvisable to ask the manufacturer to review its calibration ofthe sources to help resolve these discrepancies. With a properredundancy program to verify that the institution’s dosimetrysystem has not changed with time, there remains a small riskof error when the institution’s calibration value is used butdiffers from the manufacturer’s data.

We support the earlier AAPM recommendations onsource QA tests, their frequency, and tolerances as repro-duced here in Table I. It should be noted that the recom-mended 3% tolerance between manufacturer and institutioncalibrations discussed above applies to the mean of a batchof sources. Since individual sources may differ from themean by a greater amount, we recommend a maximum de-viation from the mean of 5% for individual sources.

For long half-life sources, the uniformity of each sourceshould be verified during the initial calibration procedure.

1568 Nath et al. : Code of practice for brachytherapy physics 1568

Medical Physics, Vol. 24, No. 10, October 1997

Page 13: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

All seed ribbons should be verified during the initial calibra-tion procedure and visually inspected to assure correct spac-ing of the seeds and the correct number of seeds. Differen-tially loaded ribbons require special consideration.

2. High strength sources for HDR applications

Currently, NIST maintains no primary air kerma strengthstandard directly applicable to HDR or PDR192Ir sources.Vendor-supplied calibration certificates are based on a vari-ety of standards the precision of which and traceability toNIST standards are often obscure. In the absence of a suit-able primary standard, the interpolative free-air secondarystandard method has become thede facto interim standardfor measurement of high intensity192Ir source strength.Briefly, this approach consists of measuring air kerma rateon the transverse axis of an HDR source at distances of 10cm–100 cm in a free-air geometry using an ion chamberwith a buildup cap thick enough to establish secondary elec-tron equilibrium at the highest photon energy encountered~about 1200 keV!. The 192Ir air kerma calibration factor isderived by interpolating between137Cs and hard orthovoltageair kerma calibration factors obtained from the NIST or anADCL calibration service. The buildup cap must be used forboth 192Ir calibration measurements and intercomparisonagainst NIST air kerma standards. Methods for interpolatingbetween the directly traceable external beam air kerma cali-bration factors to obtain an air kerma calibration factor forthe HDR 192Ir source have been reviewed by severalauthors.19–23 Room scatter corrections are generally derivedfrom the deviation of measured air kerma rate from inversesquare law19 or from shadow-block measurements.23 Addi-

tional corrections for ion recombination and photon fluencegradients across the chamber volume are applied.

The interpolative free-air secondary standard has beenimplemented by two ADCLs~K & S Associates and theUniversity of Wisconsin! and accredited by the AAPM as acalibration service. These ADCLs are authorized to calibrateusers’ re-entrant ionization chambers using this standard. In-stitutions opting to use a calibrated re-entrant ion chambershould obtain an instrument designed to high precision(,2%) in the presence of the large ion currents character-istic of this measurement with minimal ion recombinationeffects (Pion.0.98).

Specifically, the AAPM recommends the following:

~1! that a qualified medical physicist shall calibrate eachHDR/PDR source prior to clinical use in terms of airkerma strength and use this value as the basis for treat-ment planning and treatment prescription;

~2! until an appropriate primary standard is available, theinterpolative secondary free-air standard, describedabove, should be the basis of source strength determina-tion in HDR and PDR192Ir brachytherapy. Each HDRfacility should acquire a suitable re-entrant chamber, ob-tain an HDR air kerma strength calibration factor froman ADCL accredited to provide this service, and use thisinstrument for initial calibration of HDR sources.~Theapplicable recommendations of Table IV should be fol-lowed.! An acceptable but not recommended alternativeis implementation of the interpolative secondary free-standard within the institution using an appropriate ex-ternal beam ion chamber with directly traceable137Csand orthovoltage air kerma calibrations;

TABLE I. QA tests for brachytherapy sources. I, initial purchase; D, documented; and E, at every use.*

Type of source Test Frequency Tolerance

Long half-life: Physical/chemical form I DDescription Source encapsulation I D

Radionuclide distribution and source I Duniformity I 1 mmLocation of radionuclide

I 3%Long half-life: Mean of batch I 5%, DCalibration Deviation from mean E a

Calibration verificationI D

Short half-life: Physical/chemical form I DDescription Source encapsulation

E 3%Short half-life: Mean of batch E 5%Calibration Deviation from meanb E Vc

Radionuclide distribution and sourceuniformity

*Reprinted with permission from ‘‘Comprehensive QA for radiation oncology: Report of AAPM RadiationTherapy Committee Task Group 40,’’ G. J. Kutcher, L. Coia, M. Gillin, W. F. Hanson, St. Leibel, R. J.Morton, J. R. Palta, J. A. Purdy, L. E. Reinstein, G. K. Svensson, M. Weller, and L. Wingfield, Med. Phys.21,581–618~1994!. Copyright 1994 American Association of Physicists in Medicine.

aVisual check of source color code or measurement in a calibrator.bFor short half-life sources this may not always be practical.cV, visual check, autoradiograph or ionometric check.

1569 Nath et al. : Code of practice for brachytherapy physics 1569

Medical Physics, Vol. 24, No. 10, October 1997

Page 14: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

~3! following the initial calibration of an HDR or PDRsource, a confirmatory check of source strength shouldbe made by a suitable tertiary standard that is capable ofdetecting 5% errors or changes in response of the sec-ondary standard. To maximize redundancy, the tertiarystandard should utilize a different electrometer and ra-diation detector. The radiation detector can be a differention chamber or a suitable re-entrant chamber differentfrom that used for the secondary standard. This systemshould use a fixed, reproducible geometry~free-air jig ormachined phantom! that is initially calibrated against thesecondary standard.

C. Single source dosimetry data

The accuracy of dose calculations for brachytherapy im-plants is, of course, dependent on the accuracy of the dosi-metric data for the sources used. Most sources have cylindri-cal symmetry and exhibit an anisotropic dose distribution,with the dose along or near the longitudinal axis being lessthan that at the same distance along the transverse axis due toincreased filtration. Various theoretical methods rangingfrom numerical integration of point source contributions toMonte Carlo simulations appear in the literature for calculat-ing ‘‘line source’’ dose distributions. Likewise, measure-ments have been made in several ways with various types ofdetectors. Measured and calculated dose distributions aregenerally tabulated as two-dimensional arrays in either Car-tesian or polar coordinates. In this section, calculated andmeasured distributions for single sources are briefly re-viewed for137Cs sources, the high activity192Ir sources usedin high and pulsed dose rate remote afterloaders,241Amsources developed for intracavitary use,169Yb sources cur-rently being investigated for interstitial implants, and192Ir,125I, and 103Pd interstitial sources. Methods of dose calcula-tions for brachytherapy implants are briefly introduced in thenext sections.

1. ICWG formalism

The dose calculation model proposed by the ICWG hasgained wide acceptance and has been adopted by Task GroupNo. 43 of the AAPM Radiation Therapy Committee.13 Thisis a modular approach in which the effects of radionuclidedistribution within the capsule are taken into account througha geometry factorG(r ,u); the effects of absorption and scat-ter by the encapsulation and medium along the transverseaxis are taken into account by a radial dose functiong(r )and in all other directions by an angular anisotropy factorF(r ,u). Specifically, the dose rate at a pointr ,u in mediumfor a source of strengthSK is given by

D~r ,u!5LSK

G~r ,u!

G~1,p/2!F~r ,u!g~r !, ~2!

whereL5D(1,p/2)/SK is the dose rate per unit air kermastrength at 1 cm on the transverse axis. For a point sourceG(r ,u)5r 22. The ICWG recommends thatL be measuredfor each source design of each radionuclide. It is expresslyrecognized that sources of equal strengths of the same radio-

nuclide but of different designs can produce different doserates in medium. The other factors,g(r ) and F(r ,u), arealso measured or calculated in a medium. For details, thereader is referred to the AAPM Task Group No. 43 report.13

2. Sievert integral model

The Sievert integral model24,25 is the most widely usedmethod for modeling single source dose distributions around137Cs tubes and needles. This model consists of integratingthe point source dose distribution over the active length ofthe source, including corrections for photon absorption andscattering in the surrounding medium and oblique filtrationof primary photons through the source capsule. The modelrequires the user to specify the physical and active sourcelengths, the radial capsule thickness, an effective attenuationcoefficient ~sometimes called filtration coefficient!, alongwith the data required to implement the underlying isotropicpoint source model. Some algorithms model the effects ofthe finite-size active core, requiring the user to specify itsdiameter and filtration coefficient. For137Cs, the Sievertmodel has been shown to model accurately~within 5%!single source dose distributions when tested against dosemeasurements and Monte Carlo calculations.26,27 This reportrecommends that readers proceed cautiously in applying theSievert model to lower-energy sources, including192Ir wiresand seeds. Although the Sievert model accurately models thedose rate distribution near the transverse axis of192Ir, errorsin reconstructing the dose distribution near the longitudinalaxis ~where oblique filtration effects are important! as largeas 20–40% have been reported.28,29

3. Interstitial source data

Interstitial sources~seeds! for permanent and temporaryimplants are usually cylindrically shaped, a few millimetersin length, and a fraction of a millimeter in diameter. Themost common radionuclides under this category are125I,192Ir, 198Au, and 103Pd. TG-43 provides a set of data forsome designs of these sources~except gold!. These datashould be adopted by all users.F(r ,u) exhibits the anisot-ropy typical of line sources and is given as a two-dimensional table for each source. Ideally, treatment plan-ning computers should allow entry of such data tables.However, some systems treat these seeds as point sourcesproducing spherically rather than cylindrically symmetricdose distributions. In some situations this approximation issatisfactory, especially if the implant contains a large numberof seeds and/or randomly oriented seeds. For these cases, theangular anisotropy functionF(r ,u) is replaced by its 4paverage, which is referred to as the anisotropy factor. How-ever, when sources are used in linear arrays of readily deter-mined orientation, theF(r ,u) data tables should be used.

It should be noted that the Task Group No. 43 data cannotbe extended directly to include tube sources or needles. Also,several new sources such as ytterbium and americium areemerging in the field. Standard dose calculation methodsbreak down in the case of these intermediate energy photonemitters or for beta emitters that are under development for

1570 Nath et al. : Code of practice for brachytherapy physics 1570

Medical Physics, Vol. 24, No. 10, October 1997

Page 15: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

intravascular brachytherapy. A full three-dimensional matrixof dose values need to be determined for such developmentalsources. This is a nontrivial task and should be approachedas a research project.

4. Cesium intracavitary source data

Over the years, various designs of137Cs sources havebeen manufactured. Probably the most common one in use isthe 3M source. The only137Cs intracavitary source currentlyin production and clinical use is the very similarly designedCDCS-J-type source of Medi1physics, Inc. ~ArlingtonHeights, IL 60005!. Although dose distributions around thesesources can be cast into the ICWG modular formalism, theyappear in the literature in tabular form as a function of posi-tion around the source. The CDCS-J source has a slightlysmaller active length~13.5 vs 14 mm! and thinner capsule~0.5 vs 1 mm! compared to the 3M source.

For treatment planning of implants using137Cs tubes andneedles, this report recommends using single source distribu-tions calculated by the Sievert integral model until more ac-curate and complete tables, derived from direct dose mea-surements or Monte Carlo calculations, are available.Specific recommendations regarding choice of input data andpractical aspects of implementing these models are availablefrom a variety of sources.30–32 This report further recom-mends that such calculated dose distributions be carefullychecked against an appropriate benchmark prior to clinicaluse. Published dose rate tables should be used as the standardof comparison whenever available for the source type inquestion. Two-dimensional dose rate tables based on theSievert integral formalism are available for a variety of stain-less steel sheathed137Cs tubes and needles or for the many137Cs source designs used for LDR remote afterloading.These data include dosimetry applicable to the widely used3M Model 6D6C intracavitary tube33 as well as for severalobsolete source designs.27,33 Unfortunately, complete two-dimensional tables are not available for the AmershamCDCS-J source; for a partial table see Ref. 34. In these cases,the accuracy of the algorithm and the user’s understanding ofits input data should be verified by simulating a closely re-lated source design that is available in the literature. Aftervalidating the algorithm’s accuracy and implementing it forthe desired source type, the dose distribution should bechecked against manual calculations at several points. Suchchecks can be performed using the general Sievert integraltable35 or by means of the unfiltered line source formula26 inthe case of lightly filtered sources.

5. HDR and PDR iridium-192 sources

Like most cylindrical sources, the high activity192Irsource used in HDR and pulsed dose rate~PDR! remote af-terloaders exhibits an anisotropic dose distribution in water.For the HDR source, measurements in water were made byBaltas with a 0.1-cc ionization chamber36 and in polystyreneby Muller-Runkel with LiF thermoluminescent rods.37 Dosedistributions for this and the PDR source of Nucletron Cor-poration ~Columbia, MD! have been calculated by Monte

Carlo simulation.38 Measurements and calculations all had atleast 10 cm of scattering material surrounding each datapoint. For the HDR source, all three sets of data are in goodagreement except along or near the source axis, with calcu-lations showing a greater anisotropy than the measured data.Along the source axis the calculated dose rates are about15% less than measured by Baltas. The measurements ofMuller-Runkel are in better agreement with the calculationsthan with the measurements of Baltas, being within 5% ofthe former and 10% of the latter. Williamson and Li38 pub-lished two-dimensional away and along tables, as well astables of radial dose function, anisotropy functions, and doserate contours from their Monte Carlo data. Subsequent TLDand diode measurements around these sources by thisgroup39,40 have confirmed their Monte Carlo calculations. Itshould be pointed out that the data referenced above appliesto Nucletron HDR/PDR sources only. The sources used byother vendors are not the same, and data on such sources arecurrently scarce.

6. Intersource, heterogeneity, and applicatoreffects on dose

Measurements of the dose distribution around gynecologi-cal colpostat applicators containing tungsten shields havebeen made by several investigators.41–43 The goal of thesemeasurements is to obtain enough three-dimensional data ei-ther to construct look-up tables42 to be entered into treatmentplanning systems, or to derive input for simple one-dimensional dose computation algorithms.43,44 Although it isnot particularly difficult to incorporate these algorithms intoplanning computers, most commercially available systemshave no such provision. Thus the effects of the tungstenshields are generally ignored. To date, measured and calcu-lated distributions have been done for single colpostats only.Dose for an implant using two colpostats is taken as the sumof the contributions of each. This, of course, ignorescolpostat-to-colpostat shielding. Similarly, in computer dosecalculations for interstitial implants, interseed effects are ig-nored. Measurements45 and Monte Carlo calculations46 for125I implants show that for the specific geometries investi-gated the actual peripheral dose is about 6% lower than thatobtained from summing single source doses. Unlike the situ-ation with colpostats, there are as yet insufficient data torecommend incorporating interseed effects into treatmentplanning systems.

Gradually, data are becoming available to supportshielded applicator design for lower energy radionuclides.Recently, dose perturbation factors~termed ‘‘heterogeneitycorrection factors’’! down stream of small disk-shapedshields of aluminum, titanium, steel, silver, and lead placedon the transverse axes of125I, 169Yb, 192Ir, and137Cs sourceshave been measured using diode dosimetry.47 In many cases,these investigators found that these perturbation factors var-ied rapidly with the cross sectional area of the shield and itsdistance from the point of interest as well as the thickness ofthe material traversed by primary photons. This suggests thatsimple path length dose calculation algorithms44,29 cannot

1571 Nath et al. : Code of practice for brachytherapy physics 1571

Medical Physics, Vol. 24, No. 10, October 1997

Page 16: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

adequately model low-energy source dose distributions in thepresence of high density, high atomic number shields. Will-iamson et al.47 did find that if accurate three-dimensionalmodels of the source and experimental geometries wereused, Monte Carlo photon transport calculations were able toreproduce their measurements within a few per cent. Otherthan Monte Carlo simulation, no practical dose calculationalgorithms exist for accurately modeling bounded heteroge-neity effects. Convolution and scatter integration algorithms,which have the potential of greatly improved dose calcula-tion accuracy, are currently under development.48

At 125I and 103Pd photon energies, photoelectric absorp-tion contributes a larger proportion of the dose to tissue thanfor higher energies. Therefore, small variations in tissueatomic number result in significant effects on dose. The dosefrom 125I to selected tissues has been calculated49 andmeasured.50–52However, these studies involve measuring theeffects of replacing the entire water medium by muscle-,breast-, and bone-equivalent media. Recently, some datahave become available, illustrating the effects of more ana-tomically realistic bounded tissue heterogeneities surroundedby water equivalent media. Meigooniet al.53 measured theperturbation caused by a large cylinder of polystyrene in ahomogenous tissue-equivalent medium. They found that themeasured dose rates just beyond a 2-cm-thick polystyreneheterogeneity changes by as much as 130%, 55%, and 10%for 103Pd, 125I, and 241Am, respectively. In another recentstudy, Daset al.54 used TLD dosimetry to measure dose per-turbation factors for an125I source downstream of disk-shaped cavities filled with cortical bone, trabecular bone, fatsubstitute phantom, air, and lucite, and compared their mea-surements to Monte Carlo photon transport calculations.They found that the calculations and measurements agreedwithin 5%. They used the Monte Carlo technique to studythe dependence of the heterogeneity correction factor on het-erogeneity diameter, thickness, and distance from the sourceas a function of the source-to-measurement point distance.For cortical and trabecular bone, they found that the shield-ing effect varied by as much as factors of 5 and 1.33, respec-tively, with respect to distance and heterogeneity diameter,casting doubt on the utility of one-dimensional heterogeneitycorrections for this application. For the lower density hetero-geneities~air and fat!, they suggested that one-dimensionalalgorithms have an accuracy on the order of 10%. As yet,there is no model that can be used to calculate the dose to aheterogeneous medium, other than Monte Carlo simulation.However, clinical implications for125I, breast implants havebeen discussed.52

D. Source localization

Realization of the potential of brachytherapy to deliver ahigh target dose and relatively small dose to surroundingnormal tissue requires several stages of planning. One ofthese is source localization, which is the determination of thethree-dimensional coordinates and the orientation of eachsource relative to the patient anatomy. It can be accom-plished by a variety of methods, all of which require at least

two images from different perspectives. An extensive bibli-ography can be found in a recent review of the subject.55

Localization usually begins by entering source film coor-dinates into the computer by means of digitization. Eachview provides two coordinates with the rotation axis of theimager being a common coordinate. In some methods thesources must be manually matched on two films prior todigitization to be properly located within the patient. In othermethods the sources can be randomly digitized from two ormore films and the computer, using various criteria, auto-matically performs the matching. The latter are particularlyhelpful for large permanent implants with many sources, butfew treatment planning systems offer such algorithms.

It is fairly common to assign an average magnification toeach localization film, which is applied to convert film coor-dinates to patient coordinates for all the sources. If the im-plant is small and the magnification factor is that of the cen-ter of the implant, this is a reasonable approximation. Amore accurate way to convert from film to patient coordi-nates is to use a ‘‘geometric reconstruction’’ algorithm,which combines the coordinates from each view, in effect, todetermine the magnification of each source. Geometric re-construction may be used with any localization technique.For references, please see the recent review by Meli.55

A common manual matching technique uses two isocen-tric orthogonal films taken with a treatment simulator. Be-cause the anatomy is so different on orthogonal films, it issometimes difficult to match the images on the two filmscorresponding to the same source. Dummy cables with codedmarkers are sometimes helpful in correlating sources be-tween the orthogonal films. If available, the fluoroscopy ofthe simulator can be used to select films that maximizesource image matching. Sometimes orthogonal films aretaken with a portable x-ray unit. For these situations it isimportant to ensure that the films are truly orthogonal. Sometreatment planning systems combine jigs and appropriate al-gorithms to correct for any lack of orthogonality. Localiza-tion may also be accomplished from two isocentric but non-orthogonal films or from two ‘‘stereo’’ films for which thex-ray target or patient is displaced linearly between the films.Two-film techniques using an interfilm angle of less that 90°and the stereo shift method make it easier to match sourcesbecause the source image configurations are more closely thesame than on two orthogonal films. The smaller the separa-tion between the films the truer this is. However, it is alsotrue that the smaller the film separation, the poorer the local-ization accuracy. Of all techniques, orthogonal films providethe greatest accuracy because digitization errors translate tothe smallest source coordinate errors, while stereo-shift filmstypically provide the least accuracy due to poor reconstruc-tion of the depth dimension. With all techniques, digitizingaccuracy improves with increasing magnification.

Regardless of the method used, it is good practice todocument the localization accuracy. The best way to accom-plish this is, after digitization, to reconstruct source positionsfor the orientation of another film taken at the same time asthe localization films. Good agreement, best seen by overlay-ing the reconstructed source distribution on the third film, is

1572 Nath et al. : Code of practice for brachytherapy physics 1572

Medical Physics, Vol. 24, No. 10, October 1997

Page 17: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

excellent verification that the sources were properly matchedon the localization films.

The implant target is usually not identifiable on conven-tional radiographs. Thus there is no way of correlatingsources and dose distributions with the intended target vol-ume. For this reason it is becoming increasingly common toinclude imaging modalities, such as CT and MR, in thesource localization process. CT-based source localizationand dosimetry is the method of choice, except where theapplicator contains sufficient metal to cause image artifacts.The advantages are twofold:~1! the problem of matchingsources from film to film is avoided;~2! cross sectional iso-doses can be directly superimposed on the target volume andsurrounding anatomy. CT-based localization and dosimetryare particularly useful when poor quality lateral films, e.g., inthe pelvic region, hamper one’s ability to associate sourceson the anterior–posterior film with those on the lateral filmas in the case of perineal templates. Procedurally, one turnsthe gantry of the CT scanner perpendicular to the averagedirection of the needles. Slices are taken 1 cm apart corre-sponding to the 1-cm seed separations commonly used iniridium ribbons. One then assigns a seed at each needle~white spot! on each slice over the whole active length of theimplant. For head and neck cases using nylon catheters, onecan clearly see the black ‘‘holes’’ on each slice correspond-ing to the air in the catheters. For permanent seed implants,3-mm-thick slices 3 mm apart are needed to minimize thepossibility of having the same seed appear on more than oneslice. Having done this, there will still be some seeds thatappear on two adjacent slices. These seeds have to be as-signed to one slice or the other. The error in doing this arbi-trary assignment is not more than one-half the thickness ofthe slice ~1.5 mm!. Coordinates of these positions can beread off the CT console and input into the treatment planningsystem. On some treatment planning systems, one may digi-tize seed or needle locations from axial scans, one slice at atime, thus eliminating the process of entering thex,y,z co-ordinates from the keyboard. When dose distributions over-lay CT slices, doses to the target volume and critical struc-tures are easily determined and dose volume calculations canbe performed for better assessment of the implant.

For CT localization, a number of new techniques areemerging. Some of them use scout films or digitally recon-structed radiographs and offer new solutions to streaking andaliasing artifacts. Other new techniques use ultrasound orMR images. For a review, the reader is referred to the 1994AAPM summer school proceedings.56 This is a fluid areaunder active development.

Any new algorithm or a revision of an old one for local-ization of sources should be tested for accuracy using a phan-tom.

IV. IMPLANT DESIGN AND EVALUATION

A. Manual methods

The classical and traditional methods of brachytherapyplanning are available as sources of historical perspectiveand as methods of checking computer plans. In many situa-

tions these methods are valid starting points for computer-aided optimization by adaptive modification of the sourceconfiguration. When prior computer planning is not feasible,these may guide the radiation oncologist in implementation,and the physicist’s role can be to instruct and explain.

1. Manchester and Quimby systems

In the Manchester system of interstitial implantation, pe-ripheral sources define the target region and the goal is tooptimize dose uniformity.57,58 For planar and volume im-plants, planning relies on pre-calculated tables of the cumu-lated source strength per unit dose~in mg h per 1000 cGy! tobe used for adequate coverage of a given area or volume. Toobtain the total source strength, the table value is multipliedby the desired dose rate. The dose derived from the tablevalues is called the stated dose and is 10% larger than theminimum dose in the treatment region, which is the planedirectly opposite the source plane at 0.5-cm distance in thecase of planar implants and the volume enclosed by periph-eral sources in the case of volume implants. These tables arevalid only if certain source-distribution rules are followed inperforming the implant. These rules specify the fraction ofthe total source strength to be placed at the periphery, withthe remaining fraction to be distributed uniformly over theinterior. For rectangular planar implants, for example, theperipheral fraction is two-thirds if the area to be treated isless than 25 cm2, one-half if the area is between 25 cm2 and100 cm2, and one-third if the area is greater than 100 cm2.For volume implants~of any shape!, the peripheral fractionis three-fourths.

The Manchester tables were calculated for radium sourcesassuming only an inverse square attenuation of dose. Theinfluences of tissue attenuation and scatter buildup were ig-nored. For high-energy photons this is a good approximationup to about 5 cm from the source, as absorption by interven-ing tissue is canceled by in-scattering from surrounding tis-sue. Thus the tables are appropriate for other high-energyphoton emitting sources such as192Ir. However, the tablesmust not be used for low-energy emitting sources such as125Iand103Pd, which have a dose falloff considerably more rapidthan inverse square law predicts.

The Quimby implant system for interstitial implants usesequally spaced, uniform-strength sources distributed over asource plane or a treatment volume. For planar mold treat-ments, the stated dose was the maximum dose in the treat-ment plane. For Quimby volume-implant tables, the stateddose is the minimum dose within the volume. AlthoughQuimby planar mold data did not become the basis for planarinterstitial implant recommendations in the manner of theManchester system, its uniform source placement rule con-tinues to be widely applied in planar implants. It has beenshown, for volume implants,59 that uniform distribution ofsource strength leads to values of cumulated strength per unitdose that approach Manchester data ever more closely as theimplanted volume increases. Moreover, if source spacing isthe same in both directions for rectangular planar implants ofseeds in ribbons, end seeds as well as lateral-ribbon seeds

1573 Nath et al. : Code of practice for brachytherapy physics 1573

Medical Physics, Vol. 24, No. 10, October 1997

Page 18: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

may be considered peripheral, and it is found that Manches-ter placement rules are reasonably well followed, even whensource strength is distributed according to the Quimby sys-tem. These considerations make it quite feasible to useManchester data to perform approximate quality assurancechecks of computer calculated plans for Quimby-type im-plant geometries. In the case of high-dose rate remote after-loading plans that have been optimized to produce uniformdose distribution, closer agreement~within 10% for idealizedplans! may be expected.58

2. Memorial nomographs

With the advent of three-dimensional imaging techniquesthat allow more accurate assessment of resectability, fewerpermanent volume implants of unresectable tumors are beingperformed. However, for those tumors that are implantedwith 125I seeds because unresectability is determined only atthe time of surgery, a nomograph is still useful to indicatethe total seed strength required to deliver a given dose to atumor of measured dimensions and to provide some spacingguidance as well. For the nomographs developed at Memo-rial Sloan-Kettering Cancer Center,60 total seed strength is apower function of target average dimension to deliver amatched peripheral dose~MPD! of 160 Gy for average di-mensions of 3 cm or greater. The MPD method of doseevaluation by volume matching is discussed in Sec. V C be-low. The exponent~2.2! was obtained by fitting actual pa-tient MPD data. For average dimensions less than 3.0 cm, apower of 1.0 was assumed, and the coefficient was taken tobe 5 mCi ~apparent! per cm, corresponding to the originalaverage dimension rule,61 which permits the dose to increaseas the target gets smaller~proportional to the21.2 power ofaverage dimension!. The nomograph includes additionalscales to guide needle spacing for given spacings of seedsalong the needle track. A similar nomograph has been devel-oped for 103Pd seed implants.62 These nomographs are in-tended as intraoperative planning guides and should not besubstituted for more definitive planning~e.g., for prostateimplants! that uses three-dimensional images.

3. Paris system

The Paris system63,64 was developed for temporary im-plants of192Ir wire. As a planning tool, it is idealized to theextent that it assumes parallel, uniformly spaced source linesof equal length and source strength, disposed in one or moreparallel and uniformly spaced planes. In addition, since dosespecification is primarily in the central perpendicular plane,there is the requirement that the line centers fall in that plane,i.e., that the implants be rectangular in shape. For multipla-nar implants, the wire locations in transverse cross sectionshould be at the vertices either of squares or of equilateraltriangles. Linear strength density must be constant through-out the implant.

The thickness and width of the treated volume in the Parissystem are specified in the central plane, and target length isspecified in the source plane. Each dimension is consideredto be the average of individual minimum distances between

opposing undulations in the treatment isodose contour. Inorder to obtain adequate coverage with either single- ormultiple-plane implants, implantation guidelines requiresource lines to be about 50% longer than the treated lengthwhen the latter is only 4 cm and about 25% longer when thelength is 12 cm. In many cases this requirement is difficult tomeet. The ratio of the treated thickness to the source spacingfalls in the range 0.55–0.65 for planar implants, 1.55–1.60for two-plane square implants, and 1.25–1.35 for two-planetriangular implants, the ratio generally increasing withsource length and number of source lines. Extension of thetreatment width beyond the lateral source lines is about 33%of the spacing between lines for planar implants, 27% fortwo-plane square implants and 20% for two-plane triangularimplants.

Individual basal dose rates are defined in the centralplane, for planar implants, at points midway between adja-cent source lines and, for multiplanar implants, at the cen-troid of the squares or triangles formed by adjacent-sourcepenetrations. The basal dose rate for the implant is taken tobe the average of the individual basal dose rates. If the im-plantation rules have been followed carefully, the system as-sures that the isodose contour of a treatment~or reference!dose rate equal to 85% of the basal dose rate will closelyencompass the treated-volume dimensions defined above. Asa first-approximation example, planning for a single-planeimplant to treat a target volume of dimensionsL ~length!, W~width!, andT ~thickness! might proceed by choosing to usen wires, each of lengthx5(1.1311.5/L)L, spaced atd51.67T, with the number of wires given byn50.410.6(W/T).

The Paris system affords good coverage of the target vol-ume, provided implantation is accurately performed, andyields good dose uniformity within the target volume. How-ever, it includes a significant volume of normal tissue withinthe treatment isodose contour, and dose rate is adjustableonly by varying the source strength per unit distance alongsource lines.

B. Computer methods of implant design

Compared to manual methods, computer planning allowsa much better fit between achieved and desired dose levels atspecified points of clinical interest. In general, the fit is at-tained by optimizing the source configuration~positionsand/or strengths!. Adjustments of the configuration may beperformed intuitively by the planner, in which case versatileand user-friendly software is very important, or automati-cally by the computer, via algorithms that incorporate muchthe same decision criteria. In some instances the goal will beto optimize dose at points specified relative to an applicatorand, in other instances, the targeted points will be obtainedfrom three-dimensional images~CT, MR, or ultrasound! ofanatomy. Optimization may be weighted, either to avoid un-derdosing tumor or to avoid overdosing normal tissue. Theoptimization software used should indicate the goodness offit achieved, e.g., the standard deviation of the ratio ofachieved/desired doses. Thorough testing of planning soft-

1574 Nath et al. : Code of practice for brachytherapy physics 1574

Medical Physics, Vol. 24, No. 10, October 1997

Page 19: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

ware, whether obtained from commercial suppliers or devel-oped locally, is absolutely essential prior to clinical use.3

Repeat testing is required after any modification of the soft-ware.

1. Applicator-based planning

In some intracavitary and intraluminal treatments bothdose prescription points and possible source positions aredefined with respect to an applicator. For example, in high-dose rate~HDR! remote afterloading with a vaginal cylinderfor vaginal cuff treatments of post-operative endometrialcancer, the target contour is usually 0.5 cm from the cylindersurface in the upper half of the vagina and source positionsare distributed along the axis of the cylinder. Efficacioustreatment delivery depends on proper insertion and fixationof the applicator, and radiographic verification of applicatorposition is strongly recommended, but film-based planning isgenerally not required. It is at least as accurate, and certainlymore cost effective, to extract the required plan from a pre-calculated atlas of isodose rates obtained from optimizeddwell-time patterns, one for every combination of cylinderdiameter and treatment length for prescribed dose likely tobe encountered.65 One can scale dwell times for differentdoses but in practice, it is better to optimize for each doselevel because round off to the nearest second can produceslightly different times.

HDR endobronchial treatments can also be planned bypre-calculated atlases, if a constant dose is prescribed at agiven distance~usually 1 cm! from the line of source posi-tions over the entire treatment length. To limit isodose sur-face undulations to less than 2%, the prescribed distanceshould be greater than the source spacing.58 The length andlocation to be treated are decided by bronchoscopy and,again, radiographic verification is required. It is essential thatany offset between the end of the catheter or dummy sourcecable and the distal end of the treatment region be properlytaken into account.

In order to assure that the correct atlas plan has beenretrieved from the computer, it must be checked by a mem-ber of the physics staff. We recommend that hard-copy iso-dose contours be generated for review by the physician. Planparameters that need to be checked include correct applica-tor, source spacing, treatment length, stepping interval,source strength, and treatment dose. An overlay of the targetoutline on the isodose contours is frequently helpful as an aidto evaluation.

Optimization software should be used for special casesnot covered by an atlas, possibly the same software that wasused in generating the atlas. Such cases would include targetvolumes extending to the lower half of the vagina or multiplecatheter endobronchial treatments in the area of the bifurca-tion. Also, low-dose rate treatments of the abovementionedsites would not, in general, be appropriate for an atlas, be-cause of the difficulty of obtaining source strengths of pre-cisely the right relative values.

Institutional practices for intracavitary treatment of cervixcancer vary widely. When treatment points are defined with

respect to the applicator, atlases may be used to providedoses at tissue tolerance points~at markers in rectum, blad-der, etc.!. Otherwise, custom planning may be needed. Opti-mization entails trying to adjust source strengths, at radio-graphically determined positions, to bring the prescribeddose rate as close as possible to desired levels, while keepingdose rate to critical normal tissues below tolerance values. Ifoptimization by computer is performed in LDR brachy-therapy applications for the cervix, iterative optimization isrequired because only discrete source strengths areavailable66,67 whereas, when HDR is used, the essentiallycontinuous variability of source dwell time makes possibleanalytic computer optimization68–70~by least squares, for ex-ample!. It is recommended that optimization, with the aboveobjectives in mind, be performed for cervix applications,whether by an optimization algorithm or by trial and error.

2. Image-based planning

Increasingly, brachytherapy planning is based on three-dimensional images of patient anatomy. Notable examples,as already mentioned, are stereotactic temporary implants ofbrain tumors and percutaneous perineal prostate implants.

Most software used for planning stereotactic brain im-plants from CT images has been developed locally, and sev-eral approaches have been published. Some require interac-tive reconfiguration by the user, assisted by sophisticatedmanipulation and display capability,71 and others involve au-tomatic adjustments of source positions and/or strengths.72–74

In one of the latter,74 125I seed positions are iteratively least-squares optimized, first with only one seed per catheter andthen after each of a sequence of maneuvers in which nearest-neighbor catheters are combined; the combinations, whichcontinue as long as reasonable goodness-of-fit is maintained,serve both to reduce the number of skull penetrations neces-sary and to separate individual catheters enough that retainerbuttons on the surface do not interfere with one another. Forautomatic position adjustments in this type of optimization, itis essential~for convergence! that seeds be constrained not tomove outside a three-dimensional bit-map structure con-forming either to target contours drawn on the scans or, ifdesired, to smaller contours~e.g., the enhancement marginpresumed to indicate tumor!. Planning includes transformingsource coordinates planned on CT to equivalent coordinatesin the stereotactic frame system and calculating the corre-sponding angular and depth settings of the frame. It is rec-ommended that a member of the physics staff be present forthe OR procedure, to help assure that the plan is accuratelyimplemented.

Planning procedures for transperineal prostate implantsusing 125I seeds or 103Pd seeds range from CT-basedoptimization75 for fluoroscopy-guided implants toultrasound-based planning,76 or sometimes no planning at allfor ultrasound-guided implants. We recommend stronglyagainst this last option, which we believe can more likelylead to morbidity and/or underdosage. CT-based planningfacilitates localization of pubic bone and needle angulationfor better anterior coverage of large prostates, whereas

1575 Nath et al. : Code of practice for brachytherapy physics 1575

Medical Physics, Vol. 24, No. 10, October 1997

Page 20: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

ultrasound-based planning permits better definition of theprostate capsule and increases the likelihood of keepingseeds within it. One technique of needle angulation to misspubic bone involves rotation, at the template end only, of aninitially cylindrical array of needles, to produce an hourglassshape array that will be smaller in diameter in the vicinity ofthe bone than in the prostate.77 This approach, of course,requires fabrication of a custom template. Ultrasound-basedplanning generally makes use of a standard template withparallel needles, and it may be necessary to exclude patientsfor which the anterior prostate is blocked by pubic bone.Whichever imaging method is adopted, it is recommendedthat the treatment plan is designed to place seeds peripherallyto improve dose homogeneity and to avoid unnecessary ra-diation damage to the urethra.

C. Dose planning and evaluation

Whereas planning is carried out in advance of the implantprocedure for the purpose of enhancing its quality, evalua-tion is performed after the implant in order to assess qualityand to address the need for revising the source loading, treat-ment time, or written prescription. For implants performedfollowing only nomographs or other general guidelines,evaluation usually involves assigning a treatment dose basedon an analysis of isodose contours generated from radio-graphic images. Admittedly, for some types of brachy-therapy, evaluation may merge temporally with planning, asin the case of HDR remote afterloading treatments and thoseLDR treatments where sources are fixed in applicators~e.g.,eye plaques, cervix, and vaginal applicators, etc.!; for suchtreatments it is important first to be sure that the treatment isdelivered as planned and then to assess quality based on theplan.

1. Matched peripheral dose (MPD)

The MPD is defined, for permanent volume implants, asthe dose for which the contour volume equals the volume ofthe target.78 The target volume is most often approximated asthe volume of an ellipsoid having the same~orthogonal! di-mensions as the target, i.e.,V5(p/6)abc. As a dose assess-ment, MPD is an approximate method that should be usedonly for implants performed in the absence of custom plan-ning. It should no longer be used, for example, to assessprostate implant dose, for which planning is now based onthree-dimensional images. It is always an overestimate of theminimum peripheral dose, since the shapes of the matchedvolumes are never identical and, assuming geographic accu-racy, the two surfaces are interlaced, so that wherever thetarget protrudes from the treatment isodose, it protrudes to alower dose level. The extent of the overestimate, not evalu-able in the pre-CT era, has been estimated at a factor of 2, onthe average, for prostate implants, on the basis of targetsdrawn on post-implant CTs.79 However, this estimate itselfmay be an overestimate, probably due to lack of scan-to-scancontinuity in target contours, since unpublished data from thesame study also indicate a factor of 2 between the dose en-compassing 94% of the target volume and the dose encom-

passing 100%.77 In any case, it is evident that a small pro-truding spike as part of the target surface will greatlyincrease the degree of overestimate.

The MPD is linked to nomograph planning, since the totalsource strength specified by both125I and 103Pd nomographsis based on MPD data for actual implants. If moving fromnomograph planning and MPD evaluation to image-basedplanning~to achieve 100% coverage of the target volume!, itmay be advisable to lower the prescribed dose to better ap-proximate actual doses delivered historically when the MPDmethod was used. For example, if satisfactory clinical resultshad been obtained with total source strengths specified by anomograph, but post-implant MPD evaluations~from CT im-ages! consistently overestimated the minimum target-volumedose by a factor of 1.4, it could be argued that image-basedplanning should aim for a minimum dose only 70% of thenomography-planned MPD. Realistically, however, 100%coverage will seldom be achieved, and a less drastic reduc-tion in the planned minimum dose would be appropriate.Unfortunately, current data are insufficient to permit defini-tive recommendations in this instance.

2. Maximum continuous-contour dose for tumorbed implants

Planar implants~single or double plane! of 192Ir or 125Iseeds in ribbons are frequently used to treat the tumor bedafter excision of a soft tissue sarcoma. An important consid-eration is that no gaps appear between catheters in the treat-ment isodose contour.60 The assessment procedure, based onfilms taken with dummy ribbons in place, involves generat-ing isodose rate contours throughout the target region inclosely spaced~1.5–2.5 cm! planes approximately perpen-dicular to the catheter direction. Contour dose levels shouldbe no more than 20% apart, to facilitate selection within10%. The innermost continuous contour in each plane isidentified and from them the highest-dose rate that ad-equately covers the tumor is selected. Treatment time is de-termined as the quotient of the prescribed dose and the doserate selected. If wide separation of catheters in one part ofthe target region has given rise to a dose rate selection morethan 10%–15% lower than the desired dose rate~usually 10Gy/day!, it is recommended that the offending area be appro-priately hot loaded and the evaluation procedure repeated.Since there is generally a~wound healing! period of severaldays between film taking and the start of irradiation, suchadjustments and the ordering~or local assembly, for125I! ofspecial ribbon loadings are likely to be quite feasible.

3. Maximum continuous-contour dose for volumeimplants

Ideally, dose evaluation in brachytherapy should be basedon three-dimensional images of both sources and relevantanatomy, with the treatment dose specified as the dose forwhich the isodose contour just encloses the entire target vol-ume. Although we should keep trying to expand the numberof brachytherapy sites for which this ideal is approached,

1576 Nath et al. : Code of practice for brachytherapy physics 1576

Medical Physics, Vol. 24, No. 10, October 1997

Page 21: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

currently there are only a few, and we return to temporarybrain and permanent prostate implants to illustrate the con-cepts.

The quality of a stereotactic brain implant is directly re-lated to the accuracy with which planned seed positions havebeen realized. Assessment of placement accuracy within 0.3mm is readily possible, based on post-implant radiographstaken with a device, alternatively called a Lutz box or local-izer, affixed to the stereotactic frame prior to the frame’sremoval from the patient’s head.80 Anterior–posterior andlateral films each image lead-shot markers fixed at the cor-ners of a square in plastic holders on the near and far side ofthe head as well as implanted seeds~real or simulated!. Aprojective geometry algorithm due to Siddon80 enables local-ization of the seeds in the stereotactic frame system, andtheir coordinates are then transformed back to the planningCT scans via the same program that was used earlier to trans-form planned CT locations to the frame system. Individualand average ‘‘miss distances’’ can then be calculated andisodose contours plotted to check whether target coverage, atthe dose rate planned, may have been compromised. It ishighly recommended that this quality assessment procedurebe performed for each seed implant of brain, in addition toan evaluation of target coverage based on post-implant CTscans. The latter evaluation requires redrawing of target con-tours.

For permanent transperineal prostate implants, dose con-tour evaluation is possible only if post-implant scans are ob-tained, and such evaluation is strongly recommended.76,79

Seed locations should be determined directly from the CTimages, using multiple images of the same seed to improvelocalization in the longitudinal direction. An auxiliaryanterior–posterior radiograph is helpful to establish a firmseed count in case of ambiguity in the CT seed count. Iso-dose contours should be generated for overlay comparisonwith new target contours drawn by the radiation oncologist.It is important that these contours be based on the sameanatomic criteria used in defining target contours on theplanning CT images and that they not be influenced by theimages of implanted sources. It may be anticipated that thedose reported ultimately will be that which covers a givenfraction of the target volume~e.g., the 90% dose! and that, astechnological advances improve both placement accuracyand post-implant target delineation, the coverage percentageof the reported dose will increase.

4. Dose volume histograms

Although minimum dose can be approximated fairly wellfrom isodose overlays of target contours on CT scans, speci-fication of the 99% dose~the dose that covers 99% of thetarget volume!, for example, requires target-specific histo-gram data, i.e., information on what fraction of a given iso-dose contour volume falls within the target. Generation ofthis kind of data requires an algorithm that interpolates be-tween target contours to establish a three-dimensional bitmap of voxels that have, for example, values of zero insidethe target and values of one outside.74 Thus each voxel can

be assigned to a given isodose contour volume if the dosecalculated at its center is larger than the given dose and alsoassigned to the target if its bitmap element contains a zero.Since few commercially available treatment planning pro-grams currently have this feature, the AAPM can only en-courage developers to add it in the future. However, westrongly recommend that software include the capability togenerate integral~or cumulative! dose volume data and pref-erably differential volume dose data, as well. A particularvariation of the nontarget-specific differential histogram forbrachytherapy is the natural histogram, also recommended,in which the distorting influence of the inverse square law issuppressed by plotting the volume per unit23/2 power ofdose rate vs dose rate on a23/2 power scale.81 This type ofhistogram is particularly useful in assessing source configu-ration with respect to dose uniformity, on the one hand, andvolume peaking~at dose rates lower than the treatment doserate! in normal tissue, on the other.

5. Quality quantifiers

Quantities such as percentage of target volume receivinggreater than 1.5 times minimum tumor dose and percentageof normal tissue volume receiving more than 0.5 times mini-mum tumor dose are desirable. These can be achieved onlythrough a dose volume analysis based on three-dimensionalpost-implant imaging.

Dose volume data, both target specific and otherwise,have been used by a number of authors to develop implantquality assessment parameters relating to target coverage, todose uniformity, and to normal tissue irradiated. These havebeen summarized by Anderson.82 Those that are recom-mended for incorporation into brachytherapy evaluation soft-ware, in keeping with the histogram recommendationsabove, are those that do not require target-specific data; theyinclude ~1! a conformity parameter defined as the ratio oftreatment volume to target volume, as a measure of normaltissue treatment, and a uniformity parameter defined as theratio of the average dose to the prescribed dose;83 ~2! either~a! a uniformity parameter called the dose homogeneity in-dex ~DHI! and defined as the fraction of the treatment vol-ume that receives a dose between 100% and 150% of theprescribed dose;84 or ~b! a closely related uniformity param-eter called the dose nonuniformity ratio~DNR! and definedas the fraction of the treatment volume irradiated to morethan 150% of the prescribed dose;85 and ~3! uniformity andnormal-tissue-irradiation parameters based on the naturalvolume dose histogram.81 A well-designed implant may beseen as one for which the treatment dose rate~usually speci-fied, by necessity, on the basis of target coverage! corre-sponds closely to the maximum DHI, the minimum DNR, orthe lower half-maximum value of the natural histogram peak.Such correspondence assures that the target volume com-prises largely the regions of quasi-uniform dose between andamong implanted sources.

Recently, Low and Williamson86 performed an analysis ofimplant quality and found that using the dose per integrated

1577 Nath et al. : Code of practice for brachytherapy physics 1577

Medical Physics, Vol. 24, No. 10, October 1997

Page 22: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

reference air kerma~IRAK ! helped to reduce prescriptionambiguities.

D. Dose specification and reporting

A major concern among radiation oncologists practicingbrachytherapy has been the difficulty of interpreting clinicaldose response data from the literature. Although some of thisdifficulty must be attributed to the high-dose gradients foundin brachytherapy, much of it has been related to the lack ofstandardized practices of reporting dose.87 Among the sev-eral efforts to address this problem, we commend to yourattention those of the International Commission on RadiationUnits and Measurements~ICRU!88,89 and the AmericanBrachytherapy Society~ABS: formerly the American Endo-curietherapy Society!.7

1. ICRU recommendations for intracavitarybrachytherapy

Intracavitary irradiation is characterized by steep dosegradients in the vicinity of the sources and throughout thetumor and target volume. This physical characteristic, alongwith under utilization of computed tomograms~CT! andmagnetic resonance~MR! imaging techniques, makes speci-fication of target absorbed dose and maximum dose to criti-cal structures very difficult. Many quantities have been usedto quantify, prescribe, and to constrain intracavitary therapyat gynecologic malignancies including dose to point A,mgRaEq h, vaginal surface dose, and treatment time. Majorsystems for treatment of cervix cancer differ not only inchoice of dose specification criteria, but in applicator designand geometry, insertion and packing techniques, and relativeimportance of the external beam and intracavitary compo-nents of irradiation. Both the lack of a universal system ofdose specification and reporting and variation in treatmenttechniques have hampered the interpretation of data of tumorcontrol and treatment sequelae from different centers.89

The International Commission on Radiation Units andMeasurements~ICRU! has attempted to address this problemin its Report No. 38.88 In addition to reporting sourcestrengths, treatment time, and standard isodose contours~lat-eral and oblique frontal planes! the report committee recom-mends reporting~1! the dimensions of the 60-Gy isodosecontour~including external beam as well as all intracavitaryapplications!, ~2! the dose at a bladder point at the posteriorsurface of the Foley balloon on the anterior–posterior linethrough the center of the balloon,~3! the dose at the rectalpoint 0.5-cm posterior to the~opacified! vaginal cavity alongan anterior–posterior line midway between vaginal sources,~4! as defined by a lymphatic trapezoid, doses at points rep-resenting lower para-aortic as well as common and externaliliac nodes, and~5! with reference to planes tangent to theacetabula, dose at points representing distal parametrium andobturator lymph nodes.

Several recent analyses of the ICRU 60-Gy reference vol-ume have illustrated some of its weaknesses. Potish90 hasexamined the correlation of the three orthogonal ICRU iso-dose dimensions~H, W, and T!, as a function of implantdose rate, with the geometric characteristics of 90 Fletcher

intracavitary implants. His study, as well as a similar studyby Eisbruch,91 demonstrated that while the individual iso-dose dimensions were correlated with such parameters ascolpostat separation and tandem length, the most dominantparameter was mgRaEq h. Both authors showed that productof ICRU orthogonal dimensions, HWT, was uncorrelatedwith individual implant linear dimensions but highly corre-lated with mgRaEq h, for all dose levels in the therapeuticrange of interest. Eisbruch further showed that HWT was apoor predictor of the geometric volume contained within thecorresponding isodose surface and that its true volume,V(D), could be inferred with an accuracy of 5% from therelationV(D)}(mgRaEq h/D)1.64, whereD is the dose fromthe implant. These two investigations demonstrate that theparameterH•W•T, derived from the ICRU Report 38 rec-ommendations, is not a prognostic factor independent of theconcept of mgRaEq h~or equivalently IRAK!. Further, onefunction of mgRaEq h as a prescription or treatment con-straining parameter is to limit volume of tissue taken to aspecified dose by the implant.

2. Recommendations for intracavitarybrachytherapy

Partly as a result of the dosimetric limitations describedabove, intracavitary brachytherapy treatment techniques,techniques, dose prescriptions, applicator designs, andknowledge of normal-tissue and tumor dose-response rela-tionships, have evolved empirically, guided by observed con-trol and complication rates in large groups of patients treatedin a uniform fashion over many years.92 Applicator insertionremains a surgical skill, guided by palpation and direct visu-alization rather than by a quantitative geometric model of thetarget volume and surrounding normal tissues derived fromCT and MR imaging studies. It is important for the practic-ing physicist to accept that the major intracavitary brachy-therapy treatment traditions are closed systems: averageclinical outcomes for a group of patient treatments in termsof local control and complications will be predictable only ifcurrent applicator insertion and packing techniques, dosim-etric practices, and treatment prescription and loading prac-tices are consistent with evaluated base of clinical experiencefrom which the radiation oncologist’s training and knowl-edge of dose-response is derived. From this observation itfollows that a major function of the physicist is to maintainconsistency between past and current practice with respect toapplicator dosimetric characteristics and calculation of pre-scription and treatment constraining parameters such as ref-erence point doses~rectal dose, point A dose, vaginal surfacedose, etc.!. A frequently encountered problem is introductionof new sources, new applicators that differ in design fromthose previously used, and new treatment delivery technol-ogy such remote afterloading equipment. The goal is to de-velop modifications of the loading and dose prescriptionrules designed to reproduce the total dose distributionsachieved with the old equipment using the new applicatorsand sources.

To aid the physicist and radiation oncologist in maintain-

1578 Nath et al. : Code of practice for brachytherapy physics 1578

Medical Physics, Vol. 24, No. 10, October 1997

Page 23: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

ing the integrity of their treatment system, this report recom-mends the following:

~1! As recommended by the American BrachytherapySociety11 ~then American Endocurietherapy Society!,that the concept of integrated reference air kerma~IRAK ! be adopted in place of mgRaEq h or mg h as adose specification and prescription parameter in intrac-avitary brachytherapy: Integrated reference air kerma orIRAK is denoted by the symbolK ref and is defined as

Kref5(i 51

N

SK,i•t i , ~3!

where SKi is the air kerma strength of thei th source~units: cGy cm2 h21 for LDR or cGy cm2 s21 for HDR!and t I is the treatment time~units: hours for LDR andseconds for HDR! of the i th source. The recommendedunits of K ref are cGy cm2. K ref is related to mgRaEq hand mg h by

Kref5mg h•6.754, for filtration t512mm Pt,

K ref5mgRaEq h•7.227, for filtration t50.5-mm Pt.

~2! Radiation oncologists and physicists should work to-gether to develop written policies of treatment that de-fine the clinical indications for therapy, and as a functionof tumor size, location, stage, and other relevant clinicalparameters, define the external beam and brachytherapydose prescriptions that constitute the desired course oftherapy. Much mystery often surrounds the process ofmodifying the stated brachytherapy prescription to ac-commodate nonstandard tandem lengths, colpostat diam-eters, and other patient-specific parameters. For example,in mg h based treatment delivery systems, the final quan-tity of radiation delivered by a given implant, often in-volves a complex interplay between mg h, vaginal sur-face dose, rectal and bladder reference point doses,maximum time, and other parameters related to the ap-plicator dimensions and anatomic characteristics of thepatient.93,91 Treatment delivery errors and even system-atic misapplication of the system to large groups of pa-tients can result when the rules guiding individual pa-tient prescription are unspecified or known only by afew. The AAPM strongly recommends that physiciansand physicists work together to formulate prescriptionpractices in writing in as clear and straightforward afashion as possible, and tolerances for accepted devia-tions from these rules developed. Physicist review oftreatment plans and other implant calculations include anassessment of compliance with prescription rules andpolicies of treatment: deviations outside the zone of tol-erance should be reported to the radiation oncologist be-fore the completion of treatment.

~3! Conventions for radiographically localizing referencepoints, procedures for handling applicator shielding cor-rections, methods for performing associated manualtreatment time/dose calculations, optimization endpointsand methods, and any other treatment planning practices

identified as critical for maintaining the system shouldbe codified in written form and every individual treat-ment plan reviewed for compliance.

~4! Both radiation oncologists and physicists should worktogether to identify those factors which must remainconstant to maintain the consistency of the system.Physicists must come to understand that the system isabsorbed as a whole by the radiation oncologist usuallyas part of his or her training. Mixing dose specificationand treatment planning practices, applicator insertiontechniques, and dose prescriptions from different pub-lished systems should be avoided since patient responsesto treatment will not be predictable.

~5! The physicist should be involved in the process of intro-ducing technical changes in the system designed to im-prove clinical outcome. In addition to contributing tech-nical expertise to the complex process of empiricaloptimization, the physicist can ensure that desired modi-fications are consistently implemented.

3. ICRU recommendations for interstitialbrachytherapy

A draft ICRU report on dose specification in interstitialbrachytherapy has been prepared and a provisional summaryof it has appeared in print.94 On the basis of informationavailable to date, the reporting parameters therein recom-mended are closely related to those of the Paris system. Thusreported doses are defined primarily in the central plane as inthe Paris system, and the basal dose and the reference dosehave been renamed the mean central dose and the peripheraldose, respectively. Two uniformity parameters are identified:~1! the spread in the individual central doses averaged to getthe mean; and~2! the ratio of the peripheral dose to the meancentral dose. We recommend that the final report be studiedcarefully when it is published.

4. ABS recommendations for interstitialbrachytherapy

The ABS~AES at the time! physics committee, formed in1986, adopted dose specification as its first assignment andpublished Society approved recommendations in 1991.95 Theprincipal categories of information recommended for report-ing were ~1! the method of specifying target volume,whether by~in order of preference! drawing target contourson CT or MR images, by placement of surgical clips, or byprojections drawn on orthogonal radiographs,~2! a plan de-scription, including source configuration, planning method-ology ~e.g., Manchester or Paris system, Memorial nomo-graph, custom template, optimization, etc.!, and intendedtreatment and tolerance dose rates~at defined points!, and~3!evaluation of the dose distribution achieved, including speci-fication of the treatment dose and its definition~e.g., MPD,minimum dose, 99% dose, etc.!, treatment time, volume oftreatment isodose contour, with its ratio~in %! to the targetvolume, the ratio of average dose to treatment dose~as ameasure of uniformity!, and the dose at any special treatmentor tolerance points. A suggested report form, completed for

1579 Nath et al. : Code of practice for brachytherapy physics 1579

Medical Physics, Vol. 24, No. 10, October 1997

Page 24: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

four example cases, was included with the recommendations.We strongly recommend familiarity with this document andan effort to implement its recommendations.

5. AAPM recommendations for interstitialbrachytherapy

The AAPM endorses the abovementioned recommenda-tions of the ABS on dose specification and reporting of in-terstitial brachytherapy. It should also be noted the specifi-cation in terms of minimum tumor/target dose can easily leadto differences of up to a factor of 10 between prescribed andachieved dose near the boundaries of the target. Dose speci-fication remains an area of current development. Currently,regulatory definitions of misadministration in terms of a dosedeviation greater than 10% or 20% are quite meaningless inthe clinical implementation of this modality.

V. PERFORMING A BRACHYTHERAPYPROCEDURE

The following is intended to serve as a step-by-step guideto performing a brachytherapy procedure in keeping withgood medical and physics practices.

A. Initial planning

The physician–physicist interaction is a critical link inpromoting safe and accurate brachytherapy practices. Thequality of a brachytherapy procedure is dependent on thedegree to which the physicist and the physician communicatebefore, during, and after the implant. Initial planning may beas simple as scheduling a patient for a routine brachytherapyprocedure that is performed many times a month. When pos-sible, implants of a similar nature should be standardized asto sources, applicators, planning, and evaluation techniques.Even in commonly performed, uncomplicated procedures, asystematic procedure to all aspects of initial planning, appli-cator insertion, dose determination, and dose delivery shouldbe part of written procedures. For more complicated proce-dures the physicist and physician should discuss the objec-tives of the procedure and how to proceed to achieve thegoals. In either case, initial planning means that the physicistand physician have communicated about the proposed pro-cedure to ensure that both are familiar with the apparatus,have identified the target volume on an image, and agreed toan approximate isodose distribution.

The physicist should inform the physician as to the prac-tical, technical, and physics limitations inherent in a pro-posed brachytherapy case. The physicist should provide arealistic assessment of the accuracy with which the dose canbe delivered to the proposed target volume. To facilitatecommunication the planning form should include the implantobjective, the site and type of implant, apparatus needed,type and number of sources, anticipated geometry of thesources, dose to be delivered to the target volume and normaltissues, and other details of the implant.

For implants where source positions do not change fromcase to case, such as single line sources~esophagus, somebronchial, and tandem alone!, vaginal or rectal cylinders,certain templates, and some tandem and colpostat applica-

tions, standardized~library! isodoses are a reasonable alter-native to the generally preferred option of customized com-puter planning. These libraries can exist in hard-copy formor on the treatment planning computer. For more complexcases, such as some tandem and colpostat, perineal tem-plates, prostate templates,125I seed cases, and multi-planeimplants, the physician should have pre-implant images onwhich the target volume can be drawn~preferably CT scans!.A diagram of the applicator with proposed loaded sourcepositions should be prepared. In the absence of images, no-mograms or other geometrically based systems can be usedto define source loadings. In either case, initial planning in-cludes the following steps:~1! the physician identifies thetarget volume, preferably on some image, otherwise relativeto a fixed geometry applicator or by anatomical reference, orsimply by three spatial dimensions;~2! often with physicsstaff, the physician chooses the implant apparatus and sourcegeometry which suits the implant site and target volume; and~3! an approximate isodose distribution is calculated or ob-tained from a library of plans based on the physicist’s under-standing of the case.

As experience with these more complicated cases grows,rules of thumb and standardizing can be practiced to simplifythe process and enhance quality assurance. Until that expe-rience is developed, patient-specific isodose curves should begenerated based on the planned source placement. These iso-doses not only can help the physician determine the optimalsource placement but can even rule out certain applicators infavor of others. Procedures for obtaining source localizationfilms for each type of case should be well understood, as it isdifficult to re-take CT’s or radiographic films in the eventthat the first set is unusable.

B. Treatment prescription

The purpose of a treatment prescription in any area ofmedicine is to provide an unambiguous set of directions toanother person carrying out procedures on behalf of the phy-sician, such as a pharmacist filling out drug orders prescribedby a physician. For brachytherapy, the physician writing thetreatment prescription and the person performing the implantare generally the same person. Therefore, the brachytherapyphysician has the right and responsibility to modify prescrip-tions as required by further examination and new develop-ments in the clinical case. Since brachytherapy proceduresare surgical in nature, some aspects of the implant maychange from the pre-implant treatment plan because ofchanging circumstances encountered during the implant pro-cedure. Therefore, a distinction should be made between twophases of the prescription process:~1! the initial planningprior to the procedure as discussed above and~2! the post-implant treatment prescription. The pre-implant prescriptionshall be filled out before inserting radioactive sources, whichoften precedes availability of the final treatment evaluation/plan. For temporary implants, it should contain enough in-formation to guide source preparation and loading by thephysician’s designee. Normally, this would include sourcetype, source strength, batch number~where relevant!, and

1580 Nath et al. : Code of practice for brachytherapy physics 1580

Medical Physics, Vol. 24, No. 10, October 1997

Page 25: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

loading sequence/position in each catheter. The post-implantprescription shall be filled out after the evaluation process iscompleted but before the end of treatment. It should includeadditional data defining when sources are to be removed,usually including treatment time, prescribed dose~or equiva-lent parameters such as integrated reference air kerma,mg h!, and dose specification criterion.

The brachytherapy treatment prescription is a legal docu-ment, which obligates anyone associated with the case toperform duties in a way to assure that the stated details of theprescription are carried out. As such, this document shouldbe labeled ‘‘BRACHYTHERAPY PRESCRIPTIONFORM’’ in order to distinguish it from any of the othernotes, planning and summary data that may be generatedduring the treatment planning process.

Brachytherapy treatment prescriptions have similaritieswith external beam treatment prescriptions in that the doseper fraction, total dose, and treatment volume are subject tothe clinical variability inherent in the practice of medicine.On the other hand, there is a wider range of acceptable dosesin brachytherapy that provide for tumor control and yet re-spect normal tissue response tolerance. Therefore, it is clini-cally acceptable to prescribe a range of doses for a patientinstead of a single dose value. Clinical uncertainty, medicalexigency, and real life limitations mean the practice ofbrachytherapy is as much an art as other aspects of clinicalmedicine. The brachytherapy team must practice safe, highquality medicine with the patients best interest as their thera-peutic goal.

It should be noted that the AAPM recommendation ontreatment prescription is fundamentally different from theNRC definition of a ‘‘written directive.’’

C. Ordering sources

In the case where the sources needed for the implant arenot held locally in safe storage, they need to be ordered fromone of the several vendors across the country.192Ir seeds innylon ribbons,125I and 103Pd seeds are sources typically or-dered from such vendors. Regulatory agencies place manyconstraints on the ordering, possession and control, and dis-posal of radioactive sealed sources. The institution’s radio-active materials license~RML! specifies what type of sourcesand what total strength of each may be kept at any one time.If rented sources are not returned to the vendor in a timelymanner, it is possible to exceed the total strength on handpermitted by the RML. This event would then preclude onefrom ordering any more sources. With this in mind, it isrecommended that one specifies an ample strength limit foreach type of sealed source specified on the RML. Sourcescan be ordered before the implant based on the anticipatedloading arrangement, or one can wait until after the implantand computer dosimetry to order exactly what is needed. Ifdose optimization is to be achieved by using varying sourcestrengths, one can expect added complexity, source handlingconcerns, and potential confusion and error regarding theultimate location of each source strength. In the latter case,the patient may spend an additional day in the hospital while

sources are in transit. Generally, it should be possible toorder sources in advance of the implant because with expe-rience one can closely predict what will be needed~a fewextra seeds/ribbons can be ordered for good measure!.

When ordering sources for a specific patient, one shouldrequest that the patient’s name be placed on the source con-tainer, and that the sources should arrive before the day ofthe procedure~if possible!. The number of ribbons and seedsshould be specified along with the approximate strength ofeach source. For sources in nylon ribbons, the length of theplastic tip should be specified~not less than about 2 mm! aswell as the inter-source spacing~usually 1-cm center-to-center!. For any template cases, one should order coloredfilaments incorporated inside the ribbons extending from theoutermost seed to the point in the ribbon that would corre-spond to the outer end of the hollow needle. The ribbon is tobe inserted until the colored filament is just completely in-side the needle. This detects situations where the needle isclogged at the end, causing the ribbon to only appear to befully inserted. After the order for sources is placed, a logentry should be made of the patient’s name and medicalrecord number, what was ordered, and the date of the im-plant. A written procedure for ordering sources shall be pre-pared. Sources that are held in a source safe, such as cesiumtubes kept permanently and125I seeds that are stored untilused in a permanent implant, shall be inventoried every sixmonths.

D. Receiving sources

When the sources are received, one shall check that thenumber of sources and strength of each as stated on the ship-pers bill of lading agrees with what the user had ordered.Sources should be received by trained personnel~radiationsafety officer, designated staff from the radiation oncologydepartment, or radiation safety office! in a controlled andsecured area. Receiving sealed sources at the hospital receiv-ing dock for later delivery to radiation oncology is not rec-ommended.

All radioactive sources shall be stored in a lead sourcesafe of sufficient thickness to reduce the exposure rate toacceptable levels. This source safe and a working area shallbe in a secured room~hot lab!. There shall be a ‘‘CAUTION:RADIOACTIVE MATERIALS’’ sign posted on the door tothis area. Emergency instructions~including a call list ofnames and phone numbers! and a source inventory shall beposted inside the room. An individual trained in the use ofradioactive materials~usually a physicist! shall be appointedto be responsible for keeping records of the issue and returnof all sealed sources. A record shall be kept of every locationwhere sealed sources are kept and the type and approximatestrength of such sources. Remote afterloader units shall bekept in a secure location when the unit is not use. The treat-ment unit shall be posted with ‘‘CAUTION: RADIOAC-TIVE MATERIALS,’’ as well as the type and maximumstrength of the source.

When opening the source packaging, it shall be deter-mined that there is no contamination due to damage during

1581 Nath et al. : Code of practice for brachytherapy physics 1581

Medical Physics, Vol. 24, No. 10, October 1997

Page 26: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

shipping. Current regulations require that the exposure rate ata meter ~transport index, TI! and at the source containershould first be determined along with wipe testing the outercontainer. The AAPM finds this regulation to be wastefuland unnecessary. Wipe tests are not necessary at this stageand the TI verification is needed only for high-energy photonemitter shipments with a total air kerma strength exceeding50mGy m2 h21. The contents shall be examined for damageand the documentation shall be in agreement with what wasordered.

A log of the results of the exposure measurements,source-strength determination, source batch identificationnumber, and package condition shall be kept along with thepatient identification and room location. One of the primaryconcerns of the regulatory agencies is to ensure that the useris in control of radioactive material at all times. This meansthat the user should document the location of any source atany time. The logs for receipt, implantation, and shipping outcan serve as this documentation.

E. Checking sources

Now is the point in the receipt of radioactive sources toverify the vendor calibrations, as per Sec. III B.

F. Source and applicator preparation

1. Iridium-192 seeds

Often it is necessary to edit the lengths of seed ribbons.This entails cutting off one or more seeds. Editing should bedone in the hot lab behind a lead glass working area, never inthe patient’s room. Care must be taken to hold the seed to becut off with forceps to avoid having the seed fly off. Theseedited seeds should be placed in a container labeled by batchnumber and marked ‘‘RADIOACTIVE MATERIAL:192Ir’’and not stored inside the central bore of the lead carrier,which will subsequently be left in the patient’s room~it isnecessary to provide a safe area to store sources in the un-likely event that any become dislodged from the patient orapplicator!. No sources shall be left in the patient’s room thatare not part of the treatment. After preparing the ribbons, oneshall survey the area to assure that there are no stray seeds. Itis recommended that ribbons of varying lengths or activitiesbe labeled as such before being taken to the patient’s room.The ribbons should be transported to the patient’s room inthe lead shipping container inserted into a rolling cart. Thereshall be proper warning labels~‘‘CAUTION: RADIOAC-TIVE MATERIAL’’ ! affixed to the lead container that alsodescribe the source type and strength. The patient or pa-tient’s bed should be tagged as containing radioactivesources. This would be useful in the event of an emergencywhere the patient had to be removed from the room.

2. Iodine-125 and palladium-103 seeds; otherpermanently implanted seeds

Iodine seeds are received as separate seeds in small glassvials carried in lead pigs. These seeds are then loaded into‘‘magazines’’ for use in special applicators or are hand as-

sembled into source-spacer trains in sutures or plastic carri-ers. Source carrying trays shall be labeled as to source typeand strength. Mick or other seed inserters should be testedbefore being brought to the operating room. Needles andother equipment should be inspected for proper operation.This equipment can be flash sterilized near the operatingroom.

3. Cesium-137 tube sources

Tandem and colpostat source carriers shall be assembledin the hot lab. The tandem carrier commonly consists of aclear plastic tube with one end closed and a plastic insert~pusher! that has a cap at the handle end. These are bought assets where the pusher length is the same as the carrier tubelength. The total source plus spacer length is measured off onthe ‘‘pusher’’ and cut away. Care should be taken that thefinal length of the ‘‘pusher’’ keeps the source at the tip of theplastic tube with no play. The sources and spacers are in-serted into the clear tube followed by the pusher such that thecap just inserts into the carrier and makes a snug fit. Forsome of the gynecological applicators the colpostat carriersare metal ‘‘buckets’’ hinged at the end of long metal rodsdesigned to fit inside the colpostat source handles. Careshould be taken that the tube source does not fall out of thehinged bucket. The tandem and two colpostat source carriersshall be inserted into a metal carrier, which is inserted into aproperly labeled lead pig.

Prior to sterilizing, the tandem and colpostat applicatorsshould be checked for integrity, ease of operation, and fit ofall colpostat caps. It is recommended that thin lead tapemarker strips be placed around the belt of each large col-postat cap, a lead strip be run along the side of each mediumcap, and that no strip be used for small caps. This or a similarsystem allows rapid identification on localization films. Forremote afterloading cases, applicators and transfer tubesshould be checked for damage and that they connect prop-erly. A dummy source check run should be made prior totreatment for each channel to be used. This can be doneautomatically by many HDR units, otherwise a manual checkfor patency should be made.

G. Loading applicators and sources

Generally, the physicist alone should NOT load radioac-tive materials into the patient’s applicator. This task shouldbe performed by a two-member team consisting of a physi-cian and a physics staff member. A member of the physicsstaff should also be present to make survey measurements.These survey measurements shall be made with a calibratedsurvey meter and a record kept permanently either in thepatient’s chart or in separate logs, either of which may beaudited by regulatory agencies on a routine basis. A log ofwhat type and strength of sources were loaded into the pa-tient, the room number, and the date and time shall be kept.All personnel handling or assisting in the loading shall wearfilm or TLD collar badges and ring badges. A rolling leadshield or similar protective barrier shall be placed in theroom as needed to assure that no area of the hallway, adja-

1582 Nath et al. : Code of practice for brachytherapy physics 1582

Medical Physics, Vol. 24, No. 10, October 1997

Page 27: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

cent rooms, or other uncontrolled areas will exceed an expo-sure rate of 2 mR in any 1 h or more than 100 mR annuallyto any member of the general public taking into accountworkload, use factor, and occupancy factor. Protective barri-ers are not necessary for125I or 103Pd seed implants. Genericsurveys of uncontrolled area around specified rooms may bemade and kept on file. In these cases, during an actual patienttreatment, surveys of the bedside, 1 m away and in the hall-way are sufficient. It is generally recommended that leadaprons or gloves NOT be used to reduce exposure, as theseitems are ineffectual for iridium and cesium and of marginalutility for 125I and palladium ~except for high strengthsources used in temporary implants!. Time and distance con-trols should be used for radiation protection. A ‘‘CAUTION:RADIATION AREA’’ sign shall be posted on the door tothe patient’s room as well as a description of the radioactivematerial~number of ribbons, seeds, tubes, etc.! and strength,and the means to contact the RSC and physician in an emer-gency. A statement as to the time the sources were insertedand the approximate time of removal should be made in thepatient’s hospital chart. A long handle forceps and a labeledlead transport container shall be left in the room. A GeigerMuller ~GM! meter shall be readily available in case ofemergency.

With sealed sources, there is no danger of radioactivecontamination except by damage to or loss of a source. Sur-gical dressings and perineal pads should be changed onlywith the supervision of trained personnel. If a source shouldget free, it shall immediately be picked up with forceps andplaced in the lead container. The radiation safety officer andradiotherapy personnel shall be notified at once.

The nursing staff should receive in-service training atleast once per year on all aspects of radiation safety forbrachytherapy patients. This training should include theidentification of all applicators and sources, film badge pro-cedures, patient handling procedures, emergency proceduresfor whom to call, radiation exposure limits, and perspectiveson relative risks of radiation exposure versus other hazardsencountered. Nurses should have a clear idea of how muchtime at any distance they can spend with each patient.

In some cases it may be necessary to instruct the patientto not get out of bed nor assume a position that would com-promise the placement of the applicator. Nursing instructionsshould include statements similar to the following and shallbe written and placed in the patient’s chart: patient shall havea private room; nursing personnel shall wear film badges;visitors shall stay for only the time posted~provide a tablebased on total strength! at the indicated location in the room;no pregnant visitors; no visitors under 18 years of age;housekeeping may enter the room under nursing supervisionbut shall not remove anything~linen and trash is saved in theroom for survey!; a record of a dismissal survey stating thatthe there is no radiation present shall be made before patientis discharged.96

1. Iridium ribbons

Before removing any iridium ribbons from the lead con-tainer, a diagram or other system should be utilized to allow

for quick and accurate loading of all catheters or needles.One should identify previously labeled ribbons and planahead as to the order of operations. Long handled forcepsshould be used to handle the sources; however, overlong, tooheavy or otherwise cumbersome instruments that increaseloading time should not be used. Funnel-end tools should beused for loading catheters and needles if the ends are notalready funneled. There are many systems for loading rib-bons into catheters. In one of the systems, when loadingribbons into nylon catheters, one should observe that theproximal end of the source ribbon advances to the end of thecatheter or to the desired point. One should either melt orcrimp the open end closed. Needles should have stylets leftinside until loading to prevent clogging. About 1 cm of thenylon ribbon leader should protrude out from the needle foreasy removal at the end of treatment. Rubber caps should besecurely placed over the ends of the needles, providing slightpressure on the end of the ribbon. Alternatively, the exposedend of the ribbon can be folded over the end of the needleand then the rubber cap can be secured. The details in thesecond half of this paragraph are specific to a unique system.Alternate systems can be used to accomplish the same pur-pose of fixation of ribbons in the catheters.

2. Cesium tube sources

Tandem and colpostat source carriers should be securedinside the applicator by the screw-on endcaps. Care shouldbe taken not to insert a tandem source carrier too short forthe tandem, as this presents a problem both for assuring thecarrier is inserted all the way and for later removal.

3. Iodine-125 seeds and palladium-103; otherpermanently implanted seeds

A record of seeds brought to the operating room and ofthose inserted into the patient through the applicator shall bemaintained during the procedure. A survey meter with ascintillation probe designed for low-energy photon countingshould be available and should be used to verify that thereare no stray seeds in the procedure room or to find suchseeds. At the conclusion of the procedure, an accounting ofseeds used shall be performed. After the patient is removedfrom the operating room, a final survey shall be performed.A ‘‘CAUTION: RADIOACTIVE MATERIAL’’ sign shallbe attached to the patient’s bed during transport to the pri-vate room. Nursing instructions and instructions to the pa-tient upon dismissal shall be both given to the patient andcopies kept in the chart.

4. Remote afterloading

Before loading the applicator it is necessary to input pro-grammed dwell times at the console of remote afterloaders. Itis recommended that standard patterns of dwell times forsimilar applications be used whenever possible. This reducesthe chances for error in keying in dwell times and positions.A second person shall check the dwell times and positionsbefore treatment. If standard patterns are used, one personchecking the times and positions is sufficient. The physicist

1583 Nath et al. : Code of practice for brachytherapy physics 1583

Medical Physics, Vol. 24, No. 10, October 1997

Page 28: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

or trained radiation therapist should check the applicator po-sition and the connections between applicator and afterloaderhead before treatment to be sure that they agree with thetreatment plan. The radiation therapist or the physician oper-ating the console shall be trained to interpret the treatmenthistory printout during treatment in order to assure that all isproceeding correctly.

H. Removal of sources, their security, and return tothe vendor

At removal, sources shall be placed in the shielded stor-age and transport container. Sources should be counted asthey are removed. After removal, a dismissal survey shall beperformed with an appropriate detector to monitor the patientand all areas of the room. The results of this survey shall bekept on record in the radiation oncology department and beavailable for inspection by regulatory agencies. The radiationoncology department should notify nursing~and nursingshould notify housekeeping! that the room is clear. If asource of radiation is detected, it shall be located and stepsshall be taken to eliminate it. A loose source shall be pickedup with a long handled forceps or the equivalent, never withthe fingers, placed inside the shielded container, and left inthe patient’s room until the physicist clears the room. Aninvestigation of the circumstances surrounding the loosesource shall commence with the goal being to determinewhat, if any, dose reduction occurred to the target volume, ifany personnel exposure occurred, and what steps should betaken to prevent future recurrences. Reports to the NRC orother regulatory agencies are required in the event of lost orstolen sources. Sources shall be immediately returned to theshielded safe storage area after completion of treatment.Sources should be recounted in the storage area to ensure nosources have been lost in transit. An entry into the inventorylog shall be made indicating which sources, by patient, werereturned to the safe area. At this point any sources that are tobe returned to the vendor should be transferred back into theshipping lead container. Each source shall be counted as thistransfer is made to ensure that no sources are left behind, andas a final check that no sources are unaccounted for after theimplant procedure. The lead container is placed back into theouter shipping container along with document that stateswhat is being shipped back, and by whom. The exposure at 1m from the source container~transport index! shall be deter-mined. The Department of Transportation documents andshipping airbill should then be filled out and attached to thecontainer. After the package is picked up, a receipt for theitems taken shall be kept as proof of the disposition of eachpackage. A final record shall be made in the source inventorythat the lot of sources previously logged as having been re-ceived has now been returned to the vendor~specify the ven-dor and the date shipped!.

1. Permanent seed implants

A permanent implant patient maybe released from thehospital if the total exposure to any other individual from thereleased patient is unlikely to exceed 500 mR over the life of

the implant. It is permissible to include occupancy factors ofless than unity, tissue attenuation effects, and the use of localshielding in assessing compliance with this limit. The NRCis currently modifying 10 CFR 35 to conform with this rec-ommendation.

Unused seeds can be either used on a subsequent case orheld for decay~greater than ten half-lives, two years for0.5-Ci125I seeds! and then discarded. In either case, a recordof the final deposition of each seed has to kept. When de-cayed seeds are discarded the user shall deface all radioac-tive material warning labels on the source container.@CurrentNRC regulation requires that a survey of the decayed sourceswith a suitable survey meter~GM! on its most sensitive set-ting shall result in a measurement indistinguishable frombackground radiation.#

2. High-dose rate remote afterloaders

For AAPM recommendations on HDR brachytherapy, thereader is referred to the AAPM Task Group No. 59 report,which is soon to be released.

I. Source localization

Except when the geometry is completely known, as in vagi-nal cylinders for example, source localization films or otherimages are necessary in order to reconstruct in three dimen-sions the source locations for dose calculation purposes~or-thogonal films are useful in any case for documentation ofapplicator placement!. At least two images taken from dif-ferent perspectives are required. Whenever possible, CT orMRI images should be used both to locate sources and toprovide an accurate anatomical background to the dose dis-tribution. Various methods of source localization are re-viewed in Sec. III D. It is recommended that the dosimetry/physics staff supervise the localization imaging so thatfiducial marks, jigs, dummy sources, and imaging techniquesare used correctly. Films taken in the operating room shouldbe reviewed and approved before the patient is removedfrom the operating room.

J. Treatment evaluation

The planes of calculation are chosen to best represent thedose distribution relative to some anatomical structure or tothe applicator. An inappropriate choice of calculation planescan result in misinterpretation of the dose and possibly leadto a misadministration. For example, in a tandem and col-postats case, the anterior–posterior dose distribution shouldbe obtained by rotating the plane of calculation to be copla-nar with the tandem and to bisect the vaginal sources. Oth-erwise foreshortened isodose curves about the tandem willresult, giving the impression that stronger sources are re-quired at the tandem tip. Also, when orthogonal films areused for source localization, but isodoses will be computedin an axial plane in order to be superimposed on CT slices,

1584 Nath et al. : Code of practice for brachytherapy physics 1584

Medical Physics, Vol. 24, No. 10, October 1997

Page 29: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

the calculation plane must be properly oriented or the iso-dose curves that covers a particular anatomical structure maybe incorrectly chosen.

Labeling of isodose curves is another practical issue. Anysuch plot should be labeled with respect to anterior, poste-rior, superior, inferior, right, or left. Many treatment plan-ning systems are weak in their ability to label the isodoseplots automatically. Dose delivery errors can result from in-correct or inadequate labeling.

For all of the following situations, the treatment timeshould be double checked by another physicist or dosim-etrist. In addition, the localization films, source loading, iso-doses, and treatment prescription should be checked for con-sistency and accuracy.

1. Computer plan output

Where atlases are not used, customized dose distributionsare needed, which helps the physician determine the treat-ment time. In order for this information to be useful, a mini-mum set of data is needed. Dose distributions in theanterior–posterior and lateral planes or cross sections nearthe end of the implant and through the center should beobtained as a minimum. Additionally, the dose distributionshould be obtained through a plane that shows the maximumclinically significant dose that is expected to occur based onthe minimal set of plots and knowledge of the source distri-bution. Reference point doses should be obtained for criticalstructures and dose distributions should be calculatedthrough these points to determine the volume enclosed bythis dose. For geometrically irregular implants, additionaldose calculation shall be done to assess the target volumedose and any hot or cold spots that may occur.

Dose specification should be based on the recommenda-tions described in Sec. IV D. The minimum target dose,maximum clinically significant dose, and critical structuredoses shall be determined by the physician. These dosesshall be documented and any other relevant dosimetry shallbe included in the therapy chart.

2. Special considerations for HDR brachytherapy

Due to the short time frame in which HDR treatmentstake place, special attention should be given to the proce-dures used to compute the dose and dwell times. Mistakeswith HDR can occur if the physicist is pressured to performcalculations rapidly so that treatment can get started as soonas possible. One way to reduce this pressure is to use stan-dard plans whenever possible. These are sequences of dwelltimes stored in the treatment console. The physicist shouldbecome familiar enough with the type of applicator and thepossible errors involved with small deviations from the idealplacement. Upon reviewing the localization films, a decisioncan be made fairly quickly as to whether or not the applica-tion meets the geometrical requirements established for useof standards. This procedure also reduces potential errors inkeying in new dwell times for each treatment. After the pro-cedure, if there is the possibility of more than a 10% error in

target dose, one should run a computer plan for that case andmake up any differences in dose on the subsequent treat-ments.

K. Recording of physics data and other pertinentinformation in patient chart

The patient’s chart shall contain the BRACHYTHERAPYPRESCRIPTION FORM signed and dated by the physician.The prescription may include total source strength, the doseper fraction, total dose, applicator, dose rate, and total timedepending upon the clinical application~see Sec. V B!. Inaddition, the chart shall contain enough physics data so thatthe patient’s treatment can be reconstructed if the needarises. All such items inserted into the chart shall be labeledwith the patient’s name, record number, and anatomical ori-entation if applicable. The following is a list of items that thephysicist should be responsible for including in the chart:description of source type~radionuclide, active and totallength, strength!; description of applicator; source loadingpattern, spatial, and strength; individualized isodoses or ge-neric as appropriate and point doses that substantiate thephysician’s treatment prescription; orientations should be la-beled; localization films and target volume images; sourceinsertion and removal times.

We recommend that written policies and procedures bedeveloped for the following:~1! areas where sealed sourcesare stored;~2! special precautions when handling sources;~3!special instructions for nurses/housekeeping/visitors;~4!method of maintaining source accountability;~5! surveys tobe performed during treatment and at conclusion aftersources are returned to safe area~dismissal survey!; ~6! spe-cific procedures for permanent seed implants—operatingroom procedures;~7! patient room assignments and postingrequirements;~8! source loading and unloading;~9! emer-gency procedures posted and whom to contact; and~10!treatment planning and evaluation procedures for each majorsite or treatment procedure type.

VI. RECOMMENDED QUALITY ASSURANCEPROGRAM FOR BRACHYTHERAPY EQUIPMENT

A major focus of the QA program is to assure accurateoperation of all mechanical, software, and radioactive de-vices used for the planning, delivery, or QA of brachy-therapy treatments. This report divides device QA testinginto two categories:~i! acceptance testing which is per-formed upon acquiring the equipment and~ii ! periodic QAtesting. Acceptance testing is a comprehensive set of testswhich allow the physicist to evaluate the behavior and func-tion of the devices. Such testing is straightforward for simpledevices such as manual afterloading sources, but can be verycomplex and model specific in the case of remote afterload-ing units and treatment planning systems. While verificationthat the device performs as specified by the vendor is theformal endpoint of the acceptance testing, there are manyother benefits of this process. For complex systems, accep-tance testing is an opportunity for the physicist to learn indetail the operational characteristics of the system and cor-

1585 Nath et al. : Code of practice for brachytherapy physics 1585

Medical Physics, Vol. 24, No. 10, October 1997

Page 30: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

rect any false beliefs about how it works. During acceptancetesting all clinical procedures associated with device shouldbe reviewed to ensure compatibility with its operationalcharacteristics. This is also a good time to develop trainingprograms for dosimetrists, technologists, and others who willbe using the system on a daily basis. The appropriate subsetsof the acceptance testing protocol should be repeated when-ever major subsystems or components of the device are re-placed, e.g., upgrading software in a treatment planning sys-tem. Asking a new physicist to work through acceptancetesting of an unfamiliar device is an excellent method forrapidly assimilating him or her into an established brachy-therapy program.

A suggested periodic QA program for brachytherapyequipment is described in Tables II–VIII and in the follow-ing paragraphs. The AAPM views the recommended lists oftests and test frequencies not as a rigid prescription of whatmust be done, but rather as a starting for point for developinga written QA program individualized to the needs of eachinstitution. Some tests~e.g., source-strength verification! areso fundamental and universally important that the AAPMrecommends that all institutions practicing brachytherapyshall adapt them. However, in general, both clinical brachy-therapy practice standards and the demands placed on phys-

ics resources are highly variable from practice-to-practiceand from anatomic site-to-site. The AAPM believes that suchvariability in clinical practice precludes recommending asingle fixed protocol for periodic QA. Depending on the re-liability of the device in question and the clinical importanceof the target parameter, the optimal frequency of a given testmay be either smaller or larger than recommended by thisreport. Each institution practicing brachytherapy shall de-velop a written periodic QA protocol defining the tests to beperformed and their frequency for each major type of equip-ment.

A number of useful references are available for designinga program to confirm correct function of the devices andsystems used in brachytherapy. The AAPM task groups 32and 40 outline a number of basic tests and give recommen-dations as to frequency. Williamson31,97 describes modelprograms and discusses, in detail, many QA tests for low-dose rate brachytherapy~manually and remotely after-loaded!. The AAPM Task Group No. 40 report,3

Williamson,97 and the proceedings of the AAPM 1994 sum-mer school56 contain similar discussions for HDR and LDRremote afterloading technology.

A. Manual afterloading brachytherapy

Tables II and III outline core tests for manually afterload-ing sealed brachytherapy sources and applicators. Basically,the tests fall into four categories:~1! leak tests, inventories,and surveys designed to promote safety;~2! verification ofsource and applicator geometry/construction;~3! coincidenceof simulation markers and radioactive sources; and~4! accu-racy of source calibration.

The issue of leak tests illustrates a QA issue where theAAPM feels a that highly prescriptive and inflexible guide-line is inappropriate.@With the exception of one type of137Cs intracavitary tube and192Ir ribbons, NRC requires~10

TABLE II. Intracavitary source and applicator quality assurance.

Procedure End point Frequency

evaluate dimensions/serial number

source identityphysical length and diameter

initially

superposition of auto-and transmissionradiographs

active source length anduniformity, capsule thicknessaccuracy of sourceconstruction

initially

source leak test capsule integrity see texta

source calibration source strength initially, annually

dosimetric evaluationof applicator

magnitude and geometriccharacteristics of shieldingeffect

initially

orthogonal radiographsof applicators

correct source position,mechanical integrity,internal shield positioningcoincidence of dummy andradioactive source

initially, annually

measure applicatordimensions

correct diameter and length,correct diameter of allcolpostat caps and cylindersegments

initially, annually

source inventory correct source number quarterlyb

source preparation area safety of brachytherapy as neededsurvey personnel

aNRC requires leak testing, generally at 6 month intervals.bThe NRC requires~10 CFR 33.59! quarterly source inventories along withsurveys of ambient exposure rates in brachytherapy source storage areas.NRC has very detailed requirements regarding the information that must becaptured each time a sealed source is taken from or returned to its storagelocation ~10 CFR 35.406!.

TABLE III. Interstitial source and applicator quality assurance.

Procedure End point Frequency

evaluate spacing andno. seeds/ribbon

ribbon geometry and seedquantity

initially

source calibration source strength initially, each use

strength per seed orstrength per unit length

source strength uniformity initially

applicator integrity varies: initially, annuallymetal needles: sharpnessand straightnesstemplates: o-ring integrityand hole locations

evaluate dummy ribbongeometry

coincidence of dummy andradioactive sources

initially, annually

source leak test capsule integrity see text

source inventory correct source number quarterlysource preparation safety or brachytherapyarea survey personnel

1586 Nath et al. : Code of practice for brachytherapy physics 1586

Medical Physics, Vol. 24, No. 10, October 1997

Page 31: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

CFR 35.59! requires all brachytherapy sources to be leaktested at intervals of 6 months.# Many sources,137Cs intrac-avitary tubes, contain radioactivity bound in a nonsoluble,nonvolatile form which is heavily encapsulated in stainlesssteel. Unless physical inspection or history of use indicatesthe possibility of physical damage to the steel encapsulation,leak testing does not seem to be indicated at any fixed inter-val. Similarly, once released by the vendor leak testing of192Ir ribbons and HDR sources, which consist of a singlemetallic iridium–platinum alloy cylinder, is not indicated.On the other hand,125I in a volatile form is encapsulated inthin, easy-to-rupture titanium tubing for interstitial brachy-therapy. Such sources must be handled very carefully to pre-vent leakage of125I. The AAPM believes that125I seeds needto be leak tested prior to use in a second patient or after formintensive handling or manipulation.

Dosimetric evaluation of intracavitary applicators is notstraightforward as accurate measurement of brachytherapydose rates remains a research activity. However, when adopt-ing sources and applicators that differ significantly fromthose used previously, or when adopting new products forwhich published dose distributions are unavailable, carefulconsideration to its dosimetric properties should be given.Spot measurements with diode or TLD detectors, while timeconsuming, are straightforward if610% uncertainty is ac-ceptable. Alternatively, Monte Carlo photon transport calcu-lations can be used if the source/applicator geometry isknown.

Tables II–IV suggest that a manual brachytherapy QAprogram should have three testing frequencies: initial accep-tance, annual, and quarterly. Quarterly intervals are recom-

mended for isotope laboratory surveys and inventories. An-nual QA should be used as an opportunity to reviewapplicator condition and geometric integrity. Annual calibra-tion checks of all long-lived sources are recommendedmainly to confirm that the sources are correctly sorted as togroup, that source strengths have been correctly decayed, andthat the origins of intracavitary source calibrations are notlost in the sands of time and personnel changes as thesources age.

B. Remote afterloading brachytherapy devices

As with any treatment delivery system, functional remoteafterloading quality assurance tests should anticipate theprobable modes of system failure. There are three principalquality assurance end points: accuracy of the source selec-tion, spatial positioning, and control of treatment duration. Inaddition, all remote afterloaders have error and malfunctiondetection systems~‘‘interlocks’’ !, which are generally de-signed to retract all sources and sound an alarm when thetarget error condition occurs.

Specific QA procedures are dictated by individual systemdesign of which there are three major varieties:

~1! Programmable source train devices such as Nucletron’sSelectron LDR and HDR allow the user to specify theorder in which equal strength radioactive spheres of137Cs or60Co and geometrically identical spherical spac-ers are loaded into each of the treatment catheters orchannels. Different treatment times may be programmedfor each channel. These systems support only intracavi-tary therapy.

TABLE IV. Re-entrant ion chamber quality assurance.a

Procedure End point Frequency

repeated readings with lowstrength source

precision, stability initially, annually

reading/SK linear over rangeof use

linearity with respect toSK

or ion recombinationinitially, annuallyfor primary HDR sourcecalibration, ionrecombination should bemeasured each time

observe response asfunction of temperature ortime

verify that chamber is sealed orvented, depending upon design

initially, annually

reading/SK along wellaxis

spatial uniformity of response;definition of active-lengthcorrection factors

initially

reading/SK for long-livedstandard source

stability of response throughtime

initially, each use

reading/SK for each sourcetype using nist standard

define/verify calibration factors initially, two yearintervals for short-livedsources

aThe NRC has no requirements for LDR brachytherapy source calibration and places no requirements oninstruments used for same. For HDR remote afterloading systems, current NRC license review guidance~Policy and Guidance Directive FC86-4! requires source calibration to be performed by a board-certifiedphysicist.

1587 Nath et al. : Code of practice for brachytherapy physics 1587

Medical Physics, Vol. 24, No. 10, October 1997

Page 32: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

~2! Fixed source-train devices have no capability of compos-ing arbitrary source trains from elemental components~seeds, etc.!. The user can only choose which of theavailable source trains to transport from the associatedsource storage container into the remote afterloader in-termediate safe for use in subsequent treatment. Thesemachines generally have no ability to distinguish onesource from another so that loading the incorrect sourceinto the intermediate safe is always a possibility. Thesource supply for interstitial therapy is designed for pe-riodic replenishment and disposal by the user, creatingthe possibility that source order can be permuted.

~3! Single stepping source devices consist of a single cabledriven high intensity source, which moves from eachprogrammed treatment position in a catheter~dwell po-sition! after the position specific treatment time~dwelltime! has elapsed. After treating each position in a givencatheter~channel!, the source is retracted into the ma-chine, and re-injected into the next treatment catheter bymeans of a selector. Within each catheter, the insertion

depth of a dwell position sequence is continuously~ornearly so! adjustable in contrast to the programmableand fixed source-train devices, which have only a singletreatment position. Technical flexibility is enhanced bythe independently programmable dwell times, a featurethat is exploited by dwell-weight optimization algo-rithms supported by HDR/PDR treatment planningsystems.98

Single stepping source afterloaders are most commonlyused for outpatient-based HDR brachytherapy, althoughHDR intracavitary therapy can be performed with program-mable source-train devices equipped with60Co sphericalsources. One vendor has developed a stepping source remoteafterloader to support LDR brachytherapy: theMicroSelectron/PDR ~PDR refers to pulsed dose ratebrachytherapy!.99 This system simulates continuous LDRbrachytherapy by a series of mini-HDR fractions~calledpulses!, requiring 10–45 min for delivery, followed by aquiescent period. The cycle is usually repeated at hourly in-

TABLE V. Core daily quality assurance tests for a remote afterloading facility.

Test endpoint Test methodology System type

dose delivery accuracy • Verify date, time and source strength in treatment unit and planning computer. • all• Verify source strength and timer accuracy against a tertiary standard~see text!. • HDR/PDR

overall system function • Run system through a complete cycle of simulated treatment: • all- programming;- source ejection;- source retraction at end of timer countdown.

• Verify treatment status indicator lights and critical source control functions. • all• Correct function of dedicated fluoroscopy/imaging system if present. • HDR

patient/public/staff safety • Correct function:- door interlock; • HDR/PDR- area radiation monitor; • HDR/PDR- audio/visual system communication; • HDR/PDR- portable survey meter; • all- audible/visual error and alarm condition indicators; • all

• Safety equipment available: • all- emergency instructions;- emergency equipment~forceps, emergency safe, surgical supplies!;- operator’s manual;- survey meter.

• Measure hourly/weekly radiation levels after patient loaded and portable shields positioned • PDR/LDR

verify positionalaccuracy within 1 mm

Many possible tests: • all

- primary positional accuracy test for a single catheter;- deviation of ion chamber response placed near a programmed dwell position;- multiple-channel autoradiograph of every active dwell position used in the patient treatment and

compare programmed position to expected;- visually check that relative position of source tip in a ruled catheter reproduces from day-to-day.

• Autoradiograph patient-specific configuration of sources loaded into intermediate safe of device. • all fixed andprogrammablesource-train units

temporal accuracy • Many possible tests: • HDR/PDR- time duration of ‘‘source ejected’’ light;- perform a spot check of radiation output for a timed interval using tertiary calibration standard jig;- compare source arrival and departure times on printed treatment documentation with a clock or stop

watch;- for LDR, subtract treatment interruptions from overall treatment time and compare to programmed time. • LDR~optional!

1588 Nath et al. : Code of practice for brachytherapy physics 1588

Medical Physics, Vol. 24, No. 10, October 1997

Page 33: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

tervals under machine control. The dose per pulse is chosenso as to duplicate, on average, the hourly dose rate charac-teristic of the LDR treatment to be simulated.

The broad range of remote afterloader designs and theirclinical applications precludes formulation of a single onesize fits all set of QA tests: each brachytherapy physicistmust develop procedures which address the failure modescharacteristic of the specific equipment and clinical proce-dures current in his/her clinical practice. For all remote af-terloading systems, the AAPM recommends applying QA

tests at three frequencies: daily, quarterly, and annually/initially.

1. Daily remote afterloader QA protocol

For HDR brachytherapy, the daily QA routine should beexecuted before treating the first patient of the day, while foran LDR system it should be performed before initiating eachpatient treatment. Daily QA tests protocol need be performedonly on days when patients are treated, and for a multiple-

TABLE VI. Additional core quarterly quality assurance tests for a remote afterloading facility.

General endpoint Specific tests/endpoints System type

personnel safety Head/machine survey with source retracteda • allpatient safety •Important interlocks and emergency response systems function: obstructed

applicator, missing applicator, door, unlocked indexer ring, displacement, power/air pressure loss, backup battery system.

• all

•Emergency source handling tools, shielded storage container, and supplies foremergency applicator removal available and functioning.

• all

calibration of optical and pneumatic sourceposition/status detection systems; anyother preventive maintenance or inspections

•As specified by vendor. • all

correct operation of all applicators, transfertubes and source localization dummies

•Examine all dummies for kinks or bends that may shorten their axial displacementthrough applicator assembly. Check integrity of all transfer tube-applicatorinterfaces.

• all

positional accuracy: single stepping source •Verify that radioactive source position agrees with dummy marker within 0.5 mmpreviously tested against dwell position markers used in simulation.

all HDR/PDRsingle-steppingsource devices

• Confirm check cable operation.• Obtain multiple channel autoradiograph with unique dwell sequence in each

channel: verify that dwell position spacing, assignment of dwell sequence toprogrammed channel, and relative indexer length to dwell 1 are correct within1 mm.

•Confirm accuracy of daily positional test protocol.•Transfer tube length~if stability through time is not confirmed and positionalaccuracy is influenced by tube length!.

positional accuracy: multiple-sourcemachines

•Device positions source train in specified treatment location. • all

•Source trains delivered to programmed channels within 1 mm of intended location. • all•Source trains correctly sorted and composed. • programmable

source train•Source inventory correct. • all•Source trains stored in correct locations in user accessible storage location. • fixed source-train

devices

source calibration Measure source air kerma strength using a ‘secondary’ standard as described in Sec.III.

HDR/PDR

redundant source calibration checks •Difference between measured and vendor-specified air kerma strength is withinexpected margin.

• HDR/PDR

•Use tertiary source strength standard~e.g., daily/monthly output checking system!to confirm primary calibration within 5%. Different electrometerand detector to beused.

• spot check of absolute timer accuracy Various techniques available~Williamson, 1991 and 1994!. • all LDR• timer accuracy and linearity measurement • HDR/PDR

miscellaneous •Update source strength in treatment planning computer initialization file, treatmentunit and quarterly inventory.

• all

•Have a second physicist independently review the quarterly report. • HDR/PDR

aIn addition, NRC requires a complete facility survey whenever an HDR or PDR source is replaced.

1589 Nath et al. : Code of practice for brachytherapy physics 1589

Medical Physics, Vol. 24, No. 10, October 1997

Page 34: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

day LDR treatment, need be executed only once before ini-tiating the patient’s treatment, not on each day of use. Thedaily QA protocol should be designed to comprehensively, ifnonspecifically, to assess failure-prone QA endpoints of thetreatment system. Such tests should be completed before be-ginning applicator insertion in the first patient, so that anymachine malfunctions are identified before subjecting the pa-tient to any risk bearing medical procedure such as anesthe-sia. Table V lists a set of core tests, on the assumption thatthe only other routine physics QA intervals are quarterly andannual. The most useful and important tests to perform are~1! tests of overall system function~running machinethrough simulated treatment cycle! and ~2! availability ofneeded emergency and safety equipment. These tests greatly

reduce the likelihood of subjecting the patient to an unnec-essary procedure or being caught in an emergency situationwithout the resources needed to manage it.

The AAPM recommends performing some type of spotcheck of source radiation output and timer accuracy forsingle stepping high intensity source devices. A simple ap-proach is to obtain a detector reading for a fixed dwell timewith the source at a fixed location with respect to the detec-tor. For example, Williamson97 recommends a simple phan-tom that rigidly positions a Farmer chamber a short distance~1–1.5 cm! from an interstitial applicator. This tertiary cali-bration jig is calibrated against the quarterly secondary cali-bration standard. By comparing the measured charge per60-s programmed dwell time to the expected value, an over-

TABLE VII. Additional commissioning and annual quality assurance tests for a remote afterloading facility.

Test endpoint Test methodology System type

personnel andpublic safety

• Review workload and annualized unrestrictedarea/personnel exposures.

• all

• Perform facility survey if occupancy/building structurerevised.

• all

dose deliveryaccuracy

• Intercompare secondary standard used for quarterlycalibration against another departmental substandard.Obtain new calibration from ADCL if calibration morethan two years old. Perform Table III tests for re-entrantchamber if used.

• HDR/PDR

• Verify air kerma strength calibration and other annualTable I checks.

• LDR

positionalaccuracy

• Verify accuracy of any jigs or autoradiography cassettesused for daily/monthly positional accuracy verification.

• all

• Verify construction/spacing of all simulation markers~dummy sources!.

• all

• Verify position of simulation markers agrees withradioactive source for all applicator types. Verifysimulation source localization procedure.

• all

• Apply Table I/II tests to all intracavitary/interstitialapplicators.

• all

• If positional accuracy assumes fixed transfer tube length,verify length/uniformity if not checked quarterly.

• all

temporalaccuracy

• Verify timer linearity and absolute accuracy. • all

• Verify transit dose/source velocity. • all• Verify pulse sequencing. • PDR

additionalinterlock/emergencyresponse tests

• Verify that unit detects simulated detached sourcecapsule.

• HDR/PDR

• Verify emergency retraction buttons in room and manualsource retraction crank function.

• HDR/PDR

• Verify that source retracts and emergency retractionmotor activates when excessive friction/applicatorobstruction encountered by source.

• all

miscellaneous • Check that treatment unit correctly decays sourcestrengths and corrects dwell times for decay.

• all

• Review accuracy of all standard treatment configurationsstored in treatment unit.

• Review quality assurance manual and update if necessary. • all• Review compliance with personnel training requirements. • all

1590 Nath et al. : Code of practice for brachytherapy physics 1590

Medical Physics, Vol. 24, No. 10, October 1997

Page 35: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

all check of machine timer accuracy, positional accuracy,and decayed source strength can be made in a few minutes.Obviously, any reproducible detector, e.g., re-entrant cham-ber, could be used for this purpose. In general, the AAPMfeels that such daily output/timer checks are not necessaryfor conventional LDR remote afterloading systems.

The AAPM suggests including a positional accuracy testin the daily QA of all remote afterloading systems. For con-ventional fixed or programmable source-train devices, an au-toradiographic record of the source configuration prescribedfor the patient is suggested. If obtained using a properly cali-brated jig, positional accuracy can be confirmed with a pre-cision of about 1 mm. In addition, autoradiography confirmssource selection accuracy, which is essential for fixedsource-train devices for which the possibility of a permuted

source-train arrangement always exists. For HDR brachy-therapy, there are many methods of verifying positional ac-curacy~including full autoradiographic documentation of thedwell positions prescribed for each patient!. At a minimum,the AAPM suggests measuring the location of a single dwellposition and comparing it to its expected location. Recently,a simple method for detecting dwell position location using are-entrant chamber has been proposed.100

2. Quarterly remote afterloader QA protocol

The AAPM suggests a quarterly review of remote after-loader function independent of any particular patient treat-ment. A quarterly interval is suggested because this is the

TABLE VIII. Brachytherapy computer planning system quality assurance.

Function Benchmark data Frequency

verify geometric accuracy ofI/O peripherals: digitizer, CTor ultrasound interface, and plotter.

digitize/plot pattern of knowngeometry; for CT/US, imageand reconstruct phantom implant.

monthly

verify input parameters for allprecalculated single-source arrays.

published recommendations, source vendor’smechanical drawings. initially, annually

verify dose, dwell time, and treatmenttime calculations at representative pointsfor all source files.

published dose rate tables,manual calculations.

initially, annuallynew software version orsource identity

accuracy of single-source isodoses. point source output. initially, new software version

accuracy of multiple-sourceisodose contouring.

point source data forsymmetric source arrays.

initially, new software version

accuracy of plan rotationmatrix.

constancy of point doses,source positions, and isodosesunder repeated orthogonalrotations for symmetric source arrays.

initially, new software version

consistency of printed plandocumentation.

assumed input parameters. every clinical use

accuracy of coordinatereconstruction.

radiograph phantom withknown catheter geometry.

initially, new software version

accuracy of electronicdownloading of treatmentparameters of afterloader.

comparison of treatment unitand planning system printed output.

initially, new software versioneach treatment

dose-volume histogram/implantfigures of merit.

• use isotropic point sourceor segment of line sourceallowing analytic calculationof DVH.

• initially, new softwareversion

• constancy of test case DVH. • annually

optimization software. run series of test cases basedupon idealized implantgeometries of various sizes;develop a sense of whatoptimization does to animplant compared to uniformloading before trying it on patients.

initially, spot check whensoftware changes byduplicating old cases

overall system test. run series of standardizedplans to globally test allclinically used features.

initially, new softwareversion, annually

1591 Nath et al. : Code of practice for brachytherapy physics 1591

Medical Physics, Vol. 24, No. 10, October 1997

Page 36: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

frequency with which HDR sources are replaced and the fre-quency of NRC-mandated source inventory procedures forLDR. For HDR/PDR brachytherapy, the tests listed in TableVI are intended to be completed after installation of the newsource but before the device is released for patient treatment.For a conventional LDR remote afterloader, the quarterlyfrequency is suggested. Given an adequate daily QA regimenand in the absence of evidence suggesting unstable systemoperation, the AAPM does not believe that more frequentQA testing, i.e., monthly, is needed.@The AAPM recommen-dations deviate somewhat from NRC licensing requirements.NRC requires~PG&D FO86-4! monthly checks of positionalaccuracy, source calibration, timer accuracy/linearity, guidetube length constancy, and backup battery function. Surpris-ingly, NRC requires that correct placement of shields andother internal applicators be verified daily.#

Quarterly QA testing is more focused on measurement ofspecific operating characteristics, and is designed to be ex-ecuted by the physicist, in contrast to the daily tests whichmay be performed by a therapist or dosimetrist. To obtain acomprehensive sense of machine operation, the AAPM rec-ommends that the physicist perform all daily tests along withthe additional quarterly tests specified in Table VI. For HDR/PDR units, a more rigorous test of absolute timer accuracy~see Williamson97 for specific tests! should be performedalong with linearity across the dwell-time range. For LDRunits, a spot check of timer accuracy is suggested. For singlestepping source machines, positional accuracy testing is de-pendent both on the machine model and simulation sourcelocalization protocol. At the very least, the inherent posi-tional accuracy of the machine should be tested since thesource has been changed. If source positioning accuracy de-pends on transfer tube length, and this parameter is moni-tored daily, it is recommended that the length be checkedquarterly, until confidence in their geometric stability isachieved. Calibration of high intensity sources is addressedin Sec. III: Table V suggests two redundant checks be builtinto the calibration process~comparison of measured andvendor source strength and intercomparison of tertiary/dailycalibration standard and secondary standard!.

3. Acceptance testing and annual remoteafterloader QA

The annual review of remote afterloader function shouldbe comprehensive, approaching the thoroughness of initialacceptance testing in this regard. For LDR remote afterload-ers, all source and applicator tests~see Tables II and III!should be performed, including verification of sourcestrength and radiographic examination of intracavitary appli-cators. Timer accuracy and linearity should be measuredmore comprehensively~although measurement over range ofuse may be practical!. Positional accuracy should be checkedcarefully, including the condition and dimensions of alldummy simulation sources. The radioactive source locationsshould be compared directly to their dummy source counter-

parts, possibly by superposing autoradiographic images withtransmission radiographs of dummy sources on the same filmfor various types of applicators.

Additional HDR tests include comprehensive assessmentof positional accuracy, including all Table II and III posi-tional accuracy tests, measurement of transfer tube lengths,and direct verification of all simulation source localizationprotocols. Additional temporal accuracy tests include assess-ment of transit dose.101,102,97,98The AAPM Task Group No.59 is currently developing guidelines for patient-specific QAof brachytherapy suing HDR remote afterloaders.

C. Quality assurance for treatment planning andevaluation systems

Relatively little has been written on QA of clinical treat-ment planning systems in general, and even less is availablespecifically for brachytherapy treatment planning systems.These systems generally have the following components,which need to be addressed in a QA program:

~1! A method of reconstructing the three-dimensional geom-etry of the implant, consisting of a digitizer and an algo-rithm for calculating the source positions from two-dimensional projections. In addition to reconstruction fororthogonal projections or stereo-shift images, modernHDR brachytherapy software is often equipped withmore advanced features such as catheter-trajectory re-construction algorithms and a menu of algorithms fordigitizing sources from different types film geometries.Reconstruction algorithms based on CT image sets andtopograms are likely to appear in the near future.

~2! A graphics-based system for visualizing the implantedsources. Virtually all systems allow the projection of theimplant to be viewed in an arbitrarily oriented plane.Future systems are likely to permit visualization of thesources relative to soft tissue anatomy derived from CTimages.

~3! A means of assigning the source type, strength, andtreatment time~or dwell time! to each visualized source.

~4! An algorithm for calculating the absorbed dose distribu-tion given the above assignments. Currently, very simpleisotropic or filtered line source models are used to rep-resent the dose distribution from each source type. Su-perposition is then used to calculate the multiple sourcedose distribution. More modern systems allow input ofmeasured single source data and may model the effectsof applicator and tissue heterogeneities.

~5! Methods of evaluating, representing, and optimizing thedose distribution. Conventional systems require the userto heuristically evaluate the dose distribution by exam-ining isodose curve distributions in manually selectedplanes. More complex systems design for single steppingsource remote afterloaders have more advanced featuressuch as dwell-weight optimization algorithms, dose vol-

1592 Nath et al. : Code of practice for brachytherapy physics 1592

Medical Physics, Vol. 24, No. 10, October 1997

Page 37: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

ume histogram-based figures of merit for quantifying im-plant quality, and three-dimensional display of isodosesurfaces.

~6! A system producing hard-copy documentation of theplan, usually in the form of plotted isodose curves andassociated plan documentation. HDR brachytherapyplanning and evaluation systems often communicateelectronically with the treatment delivery device, elimi-nating the need to manually program the remote after-loader.

In general, brachytherapy software packages, especiallythose used for HDR treatments, are sufficiently complex thatit is impossible to test the response of the program to allpossible sequences of user input. Table VIII outlines a seriesof tests designed to verify correct function of each majorcomputational and graphic display function in relativelysimple testing situations. Many subtle input history-dependent bugs will reveal themselves only in the course ofintensive clinical use. Prevention of software related treat-ment errors, as well as data entry errors of human origin,requires careful scrutiny of each clinical treatment plan. Anindependent treatment time calculation should be performedto verify that the selected absolute absorbed dose distributionis at least approximately consistent with the specified ar-rangement, source positions, strengths, and dwell times.Williamson97 reviews a number of simple table or manualcalculation based approaches. Working through the tests de-scribed in Table VIII not only tests the software: It familiar-izes the physicist with the details of system operation andpitfalls likely to be encountered during patient planning.Planning a complex or unfamiliar type of implant can be astressful experience as one is under pressure to complete it asquickly as possible: The clinical setting is clearly not thetime to gain familiarity with and to test unfamiliar programoptions.

Dose calculation algorithms should be tested both toverify that the algorithm executes as specified~for a giveninput, the observed output is consistent with the vendor’sdescription of the algorithm! and for accuracy~algorithmoutput agrees with published reference data for the sourcetype in question!. The latter tests the user’s selection or entryof basic data from which the single source dose distributionis derived.

ACKNOWLEDGMENTS

The authors thank Deanna Jacobs for preparing and for-matting this manuscript. Many useful suggestions and a thor-ough review were provided by Michael Bohan, radiationsafety officer at Yale-New Haven Hospital.

APPENDIX A: DEFINITION OF A QUALIFIEDMEDICAL PHYSICIST

A qualified medical physicist is an individual who is com-petent to practice independently one or more of the subfieldsof medical physics. The following elements pertain to theirfields:

Therapeutic radiological physics

• therapeutic applications of x rays, gamma rays, electronand charged particle beams, neutrons, and radiations fromsealed radionuclide sources;

• equipment associated with their production, use, measure-ment, and evaluation;

• quality of images resulting from their production and use;

• medical health physics associated with this subfield.

Diagnostic radiological physics

• diagnostic applications of x rays, gamma rays from sealedsources, ultrasonic radiation, and magnetic fields;

• equipment associated with their production, use, measure-ment, and evaluation;

• quality of images resulting from their production and use;

• medical health physics associated with this subfield.

Medical nuclear physics

• therapeutic and diagnostic applications of radionuclides~except those used in sealed sources for therapeutic pur-poses!;

• equipment associated with their production, use, measure-ment, and evaluation;

• quality of images resulting from their production and use;

• medical health physics associated with this subfield.

Medical health physics

• safe use of x rays, gamma rays, electron and other chargedparticle beams, neutrons, radionuclides, and radiation fromsealed radionuclide sources for both diagnostic and thera-peutic purposes, except with regard to the application ofradiation to patients for diagnostic or therapeutic purposes;

• the instrumentation required to perform appropriate radia-tion surveys.

It is expected that an individual will not hold himself/herselfout to be qualified in a subfield for which he/she has notestablished competency. An individual will be consideredcompetent to practice one or more of the subfields of medicalphysics if that individual is certified in that subfield by any ofthe following organizations:

- The American Board of Radiology,- The American Board of Medical Physics,- The American Board of Health Physics,- The American Board of Science in Nuclear Medicine,- The Canadian College of Physicists in Medicine.

The American Association of Physicists in Medicine regards

1593 Nath et al. : Code of practice for brachytherapy physics 1593

Medical Physics, Vol. 24, No. 10, October 1997

Page 38: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

board certification, in the appropriate medical physics sub-field, and state licensure, in those states in which licensureexists, as the appropriate qualification for the designation ofqualified medical physicist.

For brachytherapy, the AAPM considers the qualified medi-cal physicist to be one who meets the above qualifications inthe subfield of therapeutic radiological physics.

In addition to the above qualifications, a qualified medicalphysicist shall meet and uphold the ‘‘Guidelines for EthicalPractice for Medical Physicists’’ as published by the Ameri-can Association of Physicists in Medicine, and satisfy statelicensure where applicable.

APPENDIX B: BRACHYTHERAPY TEAM MEMBERSAND THE RESPONSIBILITIES OF THEMEDICAL PHYSICIST

• Brachytherapy team members include:

• radiation oncologists;

• medical physicists;

• maintenance engineers;

• radiation safety officers;

• radiation safety committee chair;

• hospital administration;

• nursing;

• radiation therapists~technologists!;

• manufacturers;

• surgeons;

• primary physicians.

Brachytherapy team functions:Brachytherapy requires significant involvement and com-

munication among members of the physics, dosimetry, andmedical staff. Meticulous attention to detail and considerableinteraction among the team members is required during ap-plicator insertion, determination of dose specification pointsor volumes, prescribing dose to the tumor and normal tis-sues, computerized treatment planning, and treatment deliv-ery. The individual functions of brachytherapy team mem-bers are rooted in the training and education of the particularteam member. However, the effective and safe use ofbrachytherapy depends on a thorough understanding of thescience of all aspects of isotope treatment including the rolesof all team members.

Brachytherapy is an interactive process. The physicianneeds to be aware of dose distribution around differentsource arrangements to adequately prepare for an implant.The physicist needs to be knowledgeable of anatomy suchthat the relationships between tumor volume and surroundingnormal tissues are considered during the treatment planningprocess. This give-and-take is crucial to the planning, execu-tion, and delivery of effective and safe brachytherapy. Whenphysics is not on-site for pre-treatment planning, simulation

of source and applicator localization, source loading and un-loading, and the risk of miscommunication and treatmenterror may be increased.

Responsibilities of the medical physicist include:developing requirements and specifications for thepurchase of appropriate equipment;planning facilities to house the brachytherapy machines~including shielding design!;participating in, overseeing and monitoring facilityconstruction as needed;monitoring machine installation by the manufacturer andproviding assistance as needed;performing acceptance testing of the machine after instal-lation;commissioning the machine for clinical use;establishing methods for special clinical procedures andacquiring the necessary dosimetry data for them. Theseinclude special eye plaques, stereotactic implants, etc.;establishing procedures for treatment time calculations fortemporary brachytherapy implants;establishing methods for the determination of dose distri-butions in the patient irradiated by the brachytherapysources;participating in patient data acquisition, treatment plan-ning and implementation, and evaluation of brachytherapy treatments;implementation and monitoring of a quality assuranceprogram for personnel safety;implementation and monitoring of a quality assuranceprogram for patient safety and accuracy of dose delivery;implementation and monitoring of a maintenance sched-ule for brachytherapy equipment;development of new procedures that may lead to betterand more cost-effective use of brachytherapy in radiationoncology.

APPENDIX C: INSTRUMENTATION NEEDED FOR ABRACHYTHERAPY PHYSICS PROGRAM

Brachytherapy can be practiced with any of several tech-niques. These include manual loading of sources for intrac-avitary therapy, manual loading for interstitial therapy, re-mote afterloading for low-dose rate intracavitary orinterstitial therapy, stereotactically guided procedures fortreating brain lesions, ultrasonically guided procedures fortreating prostatic cancer, and eye plaques for treating oculartumors. Since each requires specialized equipment, a com-plete brachytherapy program is very expensive, and gener-ally confined to institutions with a large patient population.Most institutions limit their brachytherapy programs to oneor a few of the above techniques. This section lists the equip-ment needed to perform each technique adequately. The liststarts with a set of equipment for radiation safety applicableto all techniques, and necessary even if only one technique isused.

A. Equipment needed for any and all brachytherapy proce-dures:

1594 Nath et al. : Code of practice for brachytherapy physics 1594

Medical Physics, Vol. 24, No. 10, October 1997

Page 39: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

survey meter~ionization chamber type!;geiger counter with low energy probes for125I and 103Pd;preparation/storage room;lead blocks;L-blocks;forceps;well-type Nal with single channel analyzer or other equip-ment for wipe tests;calibration source for wipe test;calibrated well chamber and electrometer or ionizationchamber and jig with backup system;portable lead container;thermometer;barometer;treatment planning computer with brachytherapy soft-ware;therapy simulator or portable x-ray unit for source local-ization;mobile lead shield.

B. Equipment specifically for manually loaded intracavitaryprocedures:

cervix applicator set~tandem and colpostats!;set of cesium tube sources and dummy sources;one cesium tube source with a NIST traceable calibration;safe for storing cesium tube sources;set of Heyman sources.

C. Equipment specifically for manually loaded interstitialprocedures:

~1! permanent implants;Mick applicator kit.

~2! temporary implants;set of plastic and metal catheters and needles,templates~Syed-Neblett, perineal, etc.!,wire cutters,miscellaneous buttons, portable soldering iron, etc.

D. Equipment specifically for ultrasonically guided implants:same as C 2;ultrasound system with brachytherapy software;ultrasound transducers.

E. Equipment specifically for stereotactically guided im-plants:

head frame set;localization software;CT and/or MRI;catheter set.

F. Equipment specifically for eye plaque therapy:standard ophthalmologic instruments;gold plaque set;personal computer with eye plaque software or a calcula-tor.

G. Equipment specifically for remote afterloading:~1! low-dose rate;

LDR remote afterloader,sources,applicator sets adapted to the specific LDR unit withdummy sources and connecting tubes,portable radiographic unit,area monitors,calibration jigs,film and processor,

~2! high-dose rate;HDR source—replaced at three to four month inter-vals,HDR remote afterloader,applicator sets adapted to the specific HDR unit withdummy sources and connecting tubes,portable radiographic unit,computer, preferably with optimization software,well chamber designed or adapted for HDR or ioniza-tionchamber and electrometer,area monitors,film and processor.

APPENDIX D: MAJOR REFERENCE TO NRCDOCUMENTS REGARDING BRACHYTHERAPY

U.S. Nuclear Regulatory Commission, Title 10, Chapter1, Code of Federal Regulations-Energy, Part 20, ‘‘Standardsfor protection against radiation’’~Government Printing Of-fice, Washington, DC, 1987!.

U.S. Nuclear Regulatory Commission, Title 10, Chapter1, Code of Federal Regulations-Energy, Part 35, ‘‘Medicaluse of by-product material’’~Government Printing Office,Washington, DC, 1987!.

U.S. Nuclear Regulatory Commission,Guide for thePreparation of Applications for Medical Use Programs,Regulatory Guide 10.8, Revision 2, Washington, DC, 1987.

U.S. Nuclear Regulatory Commission,Quality Manage-ment Program and Misadministrations, Federal Register56~143!, 34 104–34 122, July 25, 1991.

U.S. Nuclear Regulatory Commission,Quality Manage-ment Program, Regulatory Guide 8.33, Washington, DC,1991.

U.S. Nuclear Regulatory Commission,Release of PatientsAfter Brachytherapy Treatment With Remote AfterloadingDevices, NRC Bulletin 93-01, U.S. Nuclear RegulatoryCommission, Washington, DC, April 20, 1993.

U.S. Nuclear Regulatory Commission,Loss of an Iridium-192 Source and Therapy Misadministration at Indiana Re-gional Cancer Center, Indiana, Pennsylvania on November16, 1992, Report NUREG-1480, U.S. Nuclear RegulatoryCommission, Washington, DC, 1993.

U.S. Nuclear Regulatory Commission,Information Re-quired for Licensing Remote Afterloading Devices, Policyand Guidance Directive FC 86-4, Washington, DC, 1994.

U.S. Nuclear Regulatory Commission,Management ofRadioactive Material Safety Programs at Medical Facilities.NUREG-1516, Washington, DC, 1996.

1595 Nath et al. : Code of practice for brachytherapy physics 1595

Medical Physics, Vol. 24, No. 10, October 1997

Page 40: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

1R. Nath, P. J. Biggs, F. J. Bova, C. C. Ling, J. A. Purdy, J. van de Geijn,and M. S. Weinhous, ‘‘AAPM code of practice for radiotherapy accelera-tors: report of AAPM Radiation Therapy Task Group No. 45,’’ Med.Phys.21, 1093–1121~1994!.

2G. P. Glasgow, J. D. Bourland, P. W. Grigsby, J. A. Meli, and K. A.Weaver,Remote Afterloading Technology, AAPM Report No. 41, Ameri-can Association of Physicists in Medicine~American Institute of Physics,New York, 1993!.

3G. J. Kutcher, L. Coia, M. Gillin, W. F. Hanson, S. Leibel, R. J. Morton,J. R. Palta, J. A. Purdy, L. E. Reinstein, G. K. Svensson, M. Weller, andL. Wingfield, Comprehensive QA for Radiation Oncology: Report ofAAPM Radiation Therapy Committee Task Group 40, AAPM Report No.46, American Association of Physicists in Medicine~American Instituteof Physics, New York, 1994!.

4American College of Radiology report,Physical Aspects of Quality As-surance~American College of Radiology, Philadelphia, 1990!.

5American College of Radiology report,Standards for Radiation Oncol-ogy ~American College of Radiology, Philadelphia, 1990!.

6American College of Medical Physics report,Radiation Control andQuality Assurance in Radiation Oncology: ACMP Report Ser. No. 2~American College of Medical Physics, Reston, VA, 1986!.

7S. Nag, A. A. Abitbol, L. L. Anderson, J. C. Blasko, A. Flores, L. B.Harrison, B. S. Hilaris, A. A. Martinez, M. P. Mehta, D. Nori, A. Porter,K. J. Rossman, B. L. Speiser, J. A. Stitt, A. M. N. Syed, and B. Vikram,‘‘Consensus guidelines for high dose rate remote brachytherapy in cervi-cal, endometrial, and endobronchial tumors,’’ Int. J. Radiat. Oncol., Biol.,Phys.27, 1241–1244~1993!.

8Inter-Society Council for Radiation Oncology report,Criteria for Radia-tion Oncology in Multidisciplinary Cancer Management~American Col-lege of Radiology, Reston, VA, 1991!.

9W. F. Hanson, ‘‘Brachytherapy source strength: Quantities, units andstandards,’’Brachytherapy Physics, edited by J. F. Williamson, B. R.Thomadsen and R. Nath~Medical Physics, Madison, WI, 1995!, pp. 71–86.

10American Association of Physicists in Medicine~AAPM!, Recommenda-tions of AAPM Task Group 32: Specification of Brachytherapy SourceStrength, AAPM Report No. 21~American Institute of Physics, NewYork, 1987!.

11J. F. Williamson, L. L. Anderson, P. W. Grigsby, A. Martinez, R. Nath,D. Neblett, A. Olch, and K. Weaver, ‘‘American Endocurietherapy Soci-ety recommendations for specification of brachytherapy source strength,’’Endocur./Hyperthermia Oncol.9, 1–7 ~1993!.

12Interstitial Collaborative Working Group~ICWG!, Interstitial Brachy-therapy: Physical, Biological, and Clinical Considerations, edited by L.L. Anderson, R. Nath, K. A. Weaver, D. Nori, T. L. Phillips, Y. H. Son,S.-T. Chiu-Tsao, A. S. Meigooni, J. A. Meli, and V. Smith~Raven, NewYork, 1990!.

13R. Nath, L. L. Anderson, G. Luxton, K. A. Weaver, J. F. Williamson, andA. S. Meigooni, ‘‘Dosimetry of interstitial brachytherapy sources: Rec-ommendations of the AAPM Radiation Therapy Committee Task GroupNo. 43,’’ Med. Phys.22, 209–234~1995!.

14T. P. Loftus, ‘‘Standardization of137Cs gamma-ray sources in terms ofexposure units~roentgens!,’’ J. Res. Natl. Bur. Stand. Sect. A74, 1–6~1970!.

15T. P. Loftus, ‘‘Standardization of192Ir gamma-ray sources in terms ofexposure,’’ J. Res. Natl. Bur. Stand.85, 15–29~1980!.

16T. P. Loftus, ‘‘Standardization of125I seeds used for brachytherapy,’’ J.Res. Natl. Bur. Stand.89, 295–303~1984!.

17H. Kubo, ‘‘Exposure contribution from TiK x rays produced in the tita-nium capsule of the clinical I-125 seed,’’ Med. Phys.12, 215–224~1985!.

18J. F. Williamson, ‘‘Monte Carlo evaluation of specific dose constants inwater for125I seeds,’’ Med. Phys.15, 686–694~1988!.

19S. J. Goetsch, F. H. Attix, D. W. Pearson, and B. R. Thomadsen, ‘‘Cali-bration of 192Ir high-dose-rate afterloading systems,’’ Med. Phys.18,462–467~1991!.

20R. K. Das, V. Mishra, H. P. Perera, A. S. Meigooni, and J. F. Williamson,‘‘A secondary air kerma strength standard for Yb-169 interstitial brachy-therapy sources,’’ Phys. Med. Biol.40, 741–756~1995!.

21F. Verhaegen, E. van Dijk, H. Thierens, A. Aalbers, and J. Seuntjens,‘‘Calibration of 192Ir brachytherapy sources in terms of reference airkerma rate with large volume spherical ionization chambers,’’ Phys. Med.Biol. 37, 2071–2082~1992!.

22T. W. M. Grimbergen and E. van Dijk, ‘‘Comparison of methods forderivation of iridium-192 calibration factors for the NE 2651 and NE2571 ionisation chambers,’’ inQuality Assurance: Activity Special ReportNo. 7 ~Nucletron-Oldelft International B. V., Veenendaal, The Nether-lands, 1995!, pp. 52–56.

23L. Beurmann, H.-M. Kramer, and H.-J. Selbach, ‘‘Reference air kermarate determination of an iridium-192 brachytherapy source,’’ inQualityAssurance: Activity Special Report No. 7~Nucletron-Oldelft InternationalB. V., Veenendaal, The Netherlands, 1995!, pp. 43–47.

24M. E. J. Young and H. F. Batho, ‘‘Dose tables for linear radium sourcescalculated by an electronic computer,’’ Br. J. Radiol.37, 38–44~1964!.

25J. F. Williamson, R. L. Morin, and F. M. Khan, ‘‘Monte Carlo evaluationof the Sievert integral for brachytherapy dosimetry,’’ Phys. Med. Biol.28, 1021–1031~1983!.

26B. L. Diffey and S. C. Klevenhagen, ‘‘An experimental and calculateddose distribution in water around CDCK-type caesium-137 sources,’’Phys. Med. Biol.20, 446–454~1975!.

27J. F. Williamson, ‘‘Monte Carlo and analytic calculation of absorbed dosenear137Cs intracavitary sources,’’ Int. J. Radiat. Oncol., Biol., Phys.15,227–237~1988!.

28M. T. Gillin, F. Lopez, R. W. Kline, D. F. Grimm, and A. Niroomand,‘‘Comparison of measured and calculated dose distributions about aniridium-192 wire,’’ Med. Phys.15, 915–918~1988!.

29J. F. Williamson, ‘‘The Sievert integral revisited: evaluation and exten-sion to 125I, 169Yb, and192Ir brachytherapy sources,’’ Int. J. Radiat. On-col., Biol., Phys.36, 1239–1350~1996!.

30BIR/IPSM. Recommendations for brachytherapy dosimetry. Report of ajoint working party of the British Institute of Radiology and the Instituteof Physical Sciences in Medicine. British Institute of Radiology, London~1993!.

31J. F. Williamson, ‘‘Practical quality assurance for low dose-rate brachy-therapy,’’ in Proceedings of an American College of Medical PhysicsSymposium on Quality Assurance in Radiotherapy Physics, edited by G.Starkshall and J. Horton~Medical Physics, Madison, WI, 1991!, pp. 139–182.

32T. J. Godden,Physical Aspects of Brachytherapy: Medical Physics Hand-book 19~Adam Hilger, Bristol, 1988!, pp. 41–76.

33V. Krishnaswamy, ‘‘Dose distributions about137Cs sources in tissue,’’Radiology15, 181–184~1978!.

34P. E. Metcalfe, ‘‘Experimental verification of cesium brachytherapy linesource emission using a semiconductor detector,’’ Med. Phys.15, 702–706 ~1988!.

35R. J. Shalek and M. Stovall, ‘‘Dosimetry in implant therapy,’’ inRadia-tion Dosimetry, Vol. III, 2nd ed., edited by F. H. Attix, W. C. Roesch, andE. Tochilin ~Academic, New York, 1969!.

36D. Baltas, R. Kramer and E. Lo¨ffler, ‘‘Measurements of anisotropy of thenew HDR iridium-192 source for the microSelectron/HDR,’’ inInterna-tional Brachytherapy Programme and Abstracts. 7th InternationalBrachytherapy Working Conference, Baltimore, MD, Veenedaal, TheNetherlands~Nucletron International B. V., 1992!, pp. 290–306.

37R. Muller-Runkel and S. H. Cho, ‘‘Anisotropy measurements of a highdose rate Ir-192 source in air and polystyrene,’’ Med. Phys.21, 1131–1134 ~1994!.

38J. F. Williamson and Z. Li, ‘‘Monte Carlo aided dosimetry of the mi-croSelectron pulsed and high dose-rate192Ir sources,’’ Med. Phys.22,809–819~1995!.

39R. K. Valicenti, A. S. Kirov, A. S. Meigooni, V. Mishra, R. K. Das, andJ. F. Williamson, ‘‘Experimental validation of Monte Carlo dose-calculations about a high-intensity Ir-192 source for pulsed dose-ratebrachytherapy,’’ Med. Phys.22, 821–830~1995!.

40A. S. Kirov, J. F. Williamson, A. S. Meigooni, and Y. Zhu, ‘‘TLD diodeand Monte Carlo dosimetry of an Ir-192 source for high dose-rate brachy-therapy,’’ Phys. Med. Biol.40, 2015–2036~1995!.

41C. C. Ling and I. J. Spiro, ‘‘Measurement of dose distribution aroundFletcher–Suit–Delcos colpostats using a Therados radiation field analyzer~RFA-3!,’’ Med. Phys.11, 326–330~1984!.

42R. Mohan, I. Y. Ding, M. K. Martel, L. L. Anderson, and D. Nori, ‘‘Mea-surements of radiation dose distributions for shielded cervical applica-tors,’’ Int. J. Radiat. Oncol., Biol., Phys.11, 861–868~1985!.

43K. J. Weeks and J. C. Dennett, ‘‘Dose calculation and measurements fora CT-compatible version of the Fletcher applicator,’’ Int. J. Radiat. On-col., Biol., Phys.18, 1191–1198~1990!.

1596 Nath et al. : Code of practice for brachytherapy physics 1596

Medical Physics, Vol. 24, No. 10, October 1997

Page 41: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

44H. Meertens and R. van der Laarse, ‘‘Screens in ovoids of a Selectroncervix applicator,’’ Radiother. Oncol.3, 69–80~1985!.

45A. S. Meigooni and R. Nath, ‘‘Interseed effects on dose for125I brachy-therapy implants,’’ Med. Phys.19, 385–390~1992!.

46G. S. Burns and D. E. Raeside, ‘‘The accuracy of single-seed dose super-position for I-125 implants,’’ Med. Phys.16, 627–631~1989!.

47J. F. Williamson, H. Perera, Z. Li, and W. R. Lutz, ‘‘Comparison ofcalculated and measured heterogeneity correction factors for125I, 137Cs,and Ir brachytherapy sources near localized heterogeneities,’’ Med. Phys.20, 209–222~1993!.

48J. F. Williamson, Z. Li, and J. W. Wong, ‘‘One-dimensional scatter-subtraction method for brachytherapy dose calculation near bounded het-erogeneities,’’ Med. Phys.20, 233–244~1993!.

49R. G. Dale, ‘‘A Monte Carlo derivation of parameters for use in the tissuedosimetry of medium and low energy nuclides,’’ Br. J. Radiol.55, 748–757 ~1982!.

50S. C. Prasad and D. A. Bassano, ‘‘Lung density effect on125I distribu-tion,’’ Med. Phys.12, 99–100~1985!.

51D. Y. C. Huang, M. C. Schell, K. A. Weaver, and C. C. Ling, ‘‘Dosedistribution of125I sources in different tissues,’’ Med. Phys.17, 826–832~1990!.

52E. D. Yorke, Y. C. D. Huang, M. C. Schell, R. Wong, and C. C. Ling,‘‘Clinical implications of I-125 dosimetry of bone and bone-soft tissueinterfaces,’’ Int. J. Radiat. Oncol., Biol., Phys.21, 1613–1619~1991!.

53A. S. Meigooni and R. Nath, ‘‘Tissue inhomogeneity correction forbrachytherapy sources in a heterogeneous phantom with cylindrical sym-metry,’’ Med. Phys.19, 401–407~1992!.

54R. K. Das, D. Keleti, Y. Zhu, A. S. Meigooni, and J. F. Williamson,‘‘Validation of Monte Carlo dose calculations near I-125 brachytherapysources in the presence of bounded heterogeneities,’’ Int. J. Radiat. On-col., Biol., Phys.38, 843–853~1997!.

55J. A. Meli, ‘‘Source localization,’’ inBrachytherapy Physics, edited by J.F. Williamson, B. R. Thomadsen, and R. Nath~Medical Physics, Madi-son, WI, 1995!, pp. 235–254.

56J. F. Williamson, B. R. Thomadsen, and R. Nath, editors,BrachytherapyPhysics~Medical Physics, Madison, WI, 1995!.

57W. J. Meredith, Ed.,Radium Dosage—the Manchester System, 2nd ed.~E. and S. Livingstone, Edinburgh, 1967!.

58L. L. Anderson and J. L. Presser, ‘‘Classical systems for temporary in-terstitial implants,’’ inModern Clinical Brachytherapy Physics, edited byJ. F. Williamson, R. Nath, and B. R. Thomadsen~Medical Physics, Madi-son, WI, 1995!.

59R. J. Shalek, M. A. Stovall, and V. A. Sampiere, ‘‘The radiation dosedistribution and dose specification in volume implants of radioactiveseeds,’’ Am. J. Roentgenol.77, 863–868~1957!.

60L. L. Anderson, K. A. Weaver, and J. A. Meli, ‘‘Physical aspects ofimplant planning,’’ in Interstitial Brachytherapy, Physical, Biological,and Clinical Considerations, edited by L. L. Anderson, R. Nath, K. A.Weaver, D. Nori, T. L. Phillips, Y. H. Son, S. T. Chiu-Tsao, A. S. Mei-gooni, J. A. Meli, and V. Smith~Interstitial Collaborative WorkingGroup! ~Raven, New York, 1990!, pp. 35–46.

61U. K. Henschke and P. Cevc, ‘‘Dimension averaging, a simple method fordosimetry of interstitial implants,’’ Radiobiol. Radiother.9, 287–298~1968!.

62L. L. Anderson, J. V. Moni, and L. B. Harrison, ‘‘A nomograph forpermanent implants of 103 Pd seeds,’’ Int. J. Radiat. Oncol., Biol., Phys.27, 129–136~1993!.

63A. Dutreix, G. Marinello, and A. Wambersie,Dosimetrie en Curiethe´ra-pie ~Masson, Paris, 1982!.

64M. T. Gillin, K. S. Albano, and B. Erickson, ‘‘Planar and volume tempo-rary interstitial implants: The Paris and other systems,’’ inModern Clini-cal Brachytherapy Physics, edited by J. F. Williamson, R. Nath, and B. R.Thomadsen~Medical Physics, Madison, WI, 1995!.

65L. L. Anderson, ‘‘Physical optimization of afterloading techniques,’’Strahlentherapie161, 264–269~1985!.

66L. M. Rosenstein, ‘‘A simple computer program for optimization ofsource loading in cervical intracavitary applicators,’’ Br. J. Radiol.50,119–122~1977!.

67L. L. Anderson, ‘‘Plan optimization and dose evaluation in brachy-therapy,’’ Semi. Radiat. Oncol.3, 290–300~1993!.

68R. S. Sloboda, ‘‘Optimization of brachytherapy dose distributions bysimulated annealing,’’ Med. Phys.19, 955–964~1992!.

69R. van der Laarse, G. K. Edmunson, R. W. Luthmann, and T. P. E. Prins,‘‘Optimization of HDR brachytherapy dose distributions,’’ Activity—TheSelectron User’s Newsletter5, 94–101~1991!.

70G. K. Edmunson, ‘‘Geometry based optimization for stepping source im-plants,’’ in Brachytherapy HDR and LDR, edited by A. A. Martinez, C.G. Orton, and R. F. Mould~Nucletron Corporation, Columbia, MD,1990!, pp. 184–192.

71K. Weaver, V. Smith, J. D. Lewis, B. Lulu, C. M. Barnett, S. A. Leibel,P. Gutin, D. Larson, and T. Phillips, ‘‘A CT-based computerized treat-ment planning system for I-125 stereotactic brain implants,’’ Int. J. Ra-diat. Oncol., Biol., Phys.18, 445–454~1990!.

72R. K. Ten Haken, R. F. Diaz, D. L. McShan, B. A. Fraass, J. A. Taren,and T. W. Hood, ‘‘From manual to 3-D computerized treatment planningfor 125-I stereotactic brain implants,’’ Int. J. Radiat. Oncol., Biol., Phys.15, 467–480~1988!.

73B. H. Bauer-Kirpes, V. Sturm, W. Schlegel, and W. J. Lorenz, ‘‘Comput-erized optimization of 1251 implants in brain tumors,’’ Int. J. Radiat.Oncol., Biol., Phys.14, 1013–1023~1988!.

74L. L. Anderson, P. J. Harrington, A. D. Osian, E. Arbit, S. A. Leibel, andM. G. Malkin, ‘‘A, versatile method for planning stereotactic brain im-plants,’’ Med. Phys.20, 1457–1464~1993!.

75J. N. Roy, S. T. Chiu-Tsao, L. L. Anderson, C. C. Ling, and K. E.Wallner, ‘‘CT-based optimized planning for transperineal prostate im-plant with customized template,’’ Int. J. Radiat. Oncol., Biol., Phys.21,483–489~1991!.

76J. C. Blasko, P. D. Grimm, and H. Ragde, ‘‘Brachytherapy and organpreservation in the management of carcinoma of the prostate,’’ Semi.Radiat. Oncol.3, 240–249~1993!.

77J. N. Roy, personal communication~1994!.78B. S. Hilaris, D. Nori, and L. L. Anderson,Atlas of Brachytherapy~Mac-

millan, New York, 1988!.79J. N. Roy, K. E. Wallner, P. J. Harrington, C. C. Ling, and L. L. Ander-

son, ‘‘A CT-based evaluation method for permanent implants: applicationto prostate,’’ Int. J. Radiat. Oncol., Biol., Phys.26, 163–169~1993!.

80R. L. Siddon and N. H. Barth, ‘‘Stereotaxic localization of intracranialtargets,’’ Int. J. Radiat. Oncol., Biol., Phys.13, 1241–1246~1987!.

81L. L. Anderson, ‘‘A ‘natural’ volume-dose histogram for brachytherapy,’’Med. Phys.13, 898–903~1986!.

82L. L. Anderson, ‘‘Dose specification and quantification of implant qual-ity,’’ in Modern Clinical Brachytherapy Physics, edited by J. F. William-son, R. Nath, and B. R. Thomadsen~Medical Physics, Madison, WI,1995!.

83J. M. Paul, P. C. Philip, R. W. Brandenburg, and F. R. Khan, ‘‘Histo-grams in brachytherapy,’’ Endocuri./Hyperther. Oncol.7, 13–26~1991!.

84A. Wu, K. Ulin, and E. S. Sternick, ‘‘A dose homogeneity index forevaluating 192Ir interstitial breast implants,’’ Med. Phys.15, 104–107~1988!.

85C. B. Saw, N. Suntharalingam, and A. Wu, ‘‘Concept of dose nonunifor-mity in interstitial brachytherapy,’’ Int. J. Radiat. Oncol., Biol., Phys.26,519–527~1993!.

86D. A. Low and J. F. Williamson, ‘‘The evaluation of optimized implantsfor idealized implant geometries,’’ Med. Phys.22, 1477–1485~1995!.

87A. J. Olch, A. R. Kagan, M. Wollin, S. Chan, and J. Bellotti, ‘‘Multi-institutional survey of techniques in volume iridium implants,’’Endocuri./Hyperther. Oncol.2, 193–197~1986!.

88D. Chassagne, A. Dutreix, P. Almond, J. M. V. Burgers, M. Busch, andC. A. Joslin,Dose and Volume Specification for Reporting IntracavitaryTherapy in Gynecology, ICRU Report No. 38~International Commissionon Radiation Units and Measurements, Bethesda, MD, 1985!.

89B. A. Esche, J. M. Crook, J. Isturiz, and J.-C. Horiot, ‘‘Reference volume,milligram-hours and external irradiation for the Fletcher applicator,’’ Ra-diother. Oncol.9, 255–2611~1987!.

90R. A. Potish, ‘‘The effect of applicator geometry on dose specification incervical cancer,’’ Int. J. Radiat. Oncol., Biol., Phys.18, 1513–1520~1990!.

91A. Eisbruch, J. F. Williamson, R. Dickson, P. W. Grigsby, and C. A.Perez, ‘‘Estimation of tissue volume irradiated by intracavitary im-plants,’’ Int. J. Radiat. Oncol., Biol., Phys.25, 733–744~1993!.

92C. A. Perez, D. M. Garcia, P. W. Grigsby, and J. F. Williamson, ‘‘Clini-cal applications of brachytherapy,’’ inPrinciples and Practice of Radia-tion Oncology, 2nd ed., edited by C. A. Perez and L. W. Brady~J. B.Lippincott, Philadelphia, 1992!, pp. 300–367.

1597 Nath et al. : Code of practice for brachytherapy physics 1597

Medical Physics, Vol. 24, No. 10, October 1997

Page 42: Code of practice for brachytherapy physics: Report of the ... · Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56 Ravinder

93G. H. Fletcher and A. D. Hamberger, ‘‘Squamous cell carcinoma of theuterine cervix,’’ in Textbook of Radiotherapy, edited by G. H. Fletcher~Lea & Fibiger, Philadelphia, 1980!, pp. 720–772.

94W. F. Hanson and M. Graves, ‘‘ICRU recommendations on dose speci-fication in brachytherapy,’’ inModern Clinical Brachytherapy Physics,edited by J. F. Williamson, R. Nath, and B. R. Thomadsen~MedicalPhysics, Madison, WI, 1995!.

95L. L. Anderson, R. Nath, A. J. Olch, and J. Roy, ‘‘American Endocuri-etherapy Society recommendations for dose specification in brachy-therapy,’’ Endocuri./Hyperther. Oncol.7, 1–12~1991!.

96M. A. Breeding and M. Wollin, ‘‘Working Safely Around ImplantedRadiation Sources,’’ Nursing6, 58–63~1976!.

97J. F. Williamson, G. A. Ezzell, A. J. Olch, and B. R. Thomadsen, ‘‘Qual-ity assurance for high dose rate brachytherapy,’’ inHigh Dose RateBrachytherapy: A Textbook, edited by S. Nag~Futura, Armonk, NY,1994!, pp. 147–212.

98G. A. Ezzell and R. W. Luthmann, ‘‘Clinical implementation of dwelltime optimization techniques for single stepping-source remote applica-

tors,’’ in Brachytherapy Physics, edited by J. F. Williamson, B. R. Tho-madsen, and R. Nath~Medical Physics, Madison, WI, 1995!, pp. 617–639.

99J. F. Williamson, P. W. Grigsby, A. S. Meigooni, and S. Teague, ‘‘Clini-cal physics of pulsed dose rate remotely-afterloaded brachytherapy,’’ inBrachytherapy Physics, edited by J. F. Williamson, B. R. Thomadsen,and R. Nath~Medical Physics, Madison, WI, 1995!, pp. 577–616.

100L. A. De Werd, P. Jursinic, R. Kitchen, and B. R. Thomadsen, ‘‘Qualityassurance tool for high dose rate brachytherapy,’’ Med. Phys.22, 435–440 ~1995!.

101K. T. Bastin, M. S. Podgorsak, and B. R. Thomadsen, ‘‘The transit dosecomponent of high dose-rate brachytherapy: Direct measurements andclinical implications,’’ Int. J. Radiat. Oncol., Biol., Phys.26, 695–702~1993!.

102P. V. Houdek, J. G. Schwade, X. Wu, A. Pisciotta, J. A. Fiedler, C. F.Serago, A. M. Markoe, A. A. Abitbol, A. A. Levin, P. G. Brauhsch-weiger, and M. D. Sklar, ‘‘Dose determination in high dose-rate brachy-therapy,’’ Int. J. Radiat. Oncol., Biol., Phys.24, 795–801~1992!.

1598 Nath et al. : Code of practice for brachytherapy physics 1598

Medical Physics, Vol. 24, No. 10, October 1997