Vision 2020 Collection
Vision 2020 Collection
Vision 2020 Collection
Received 19 January 2007; revised 7 March 2007; accepted for publication 15 March 2007;
published 17 April 2007
A modern approach to quality was developed in the United States at Bell Telephone Laboratories
during the first part of the 20th century. Over the years, those quality techniques have been adopted
and extended by almost every industry. Medicine in general and radiation oncology in particular
have been slow to adopt modern quality techniques. This work contains a brief description of the
history of research on quality that led to the development of organization-wide quality programs
such as Six Sigma. The aim is to discuss the current approach to quality in radiation oncology as
well as where quality should be in the future. A strategy is suggested with the goal to provide a
threshold improvement in quality over the next 10 years. 2007 American Association of Physicists in Medicine. DOI: 10.1118/1.2727748
Key words: quality, variation, process improvement, quality management
I. BACKGROUND AND INTRODUCTION
The industrial revolution resulted in notable worldwide contributions including mass production. In the early part of the
20th century, the emphasis was to bring as much product to
market as possible in the shortest period of time, often with
little attention to quality. The primary method to ensure quality was by inspection. Each component that created the final
product was inspected to verify it was within the specifications set by product engineers. If a component was outside
specifications, then it was either scrapped or sent back for
rework and subsequent reinspection.
During this period, American Telephone & Telegraph
AT&T was hard at work building facilities to support a
universal telephone service. Western Electric Company created by AT&T as a constant source of supply was struggling
with problems of mass production, including the interchange
of parts, precision, and reliability.1 Scientists and engineers
at Bell Telephone Laboratories Bell Labs, a subsidiary of
AT&T, were engaged in product research as well as research
on problems of quality associated with mass production. A
major emphasis was on the application of probability and
statistics to quality.
By the mid 1940s, significant progress was being made on
new approaches to quality. Researchers at Bell Labs realized
that product inspection was not adequate to ensure a highquality product.1 It was much more important to control the
variation in the process that created the product. A significant
invention was the control chart where data acquired by inspection was plotted sequentially to characterize process
variation.2 Based in part on this work, W. Edwards Deming,
Joseph M. Juran, Kaoru Ishikawa, and others were creating
new approaches to quality that extended beyond the manufactured product.3 Unfortunately for AT&T, much of this research did not translate into their production efforts.
After entering World War II in December 1941, the U.S.
enacted legislation to gear the civilian economy to enhance
military production. The armed forces also helped suppliers
by sponsoring training courses on ways to improve quality.
1529
0094-2405/2007/345/1529/6/$23.00
1529
1530
1530
TABLE I. Seven basic tools of quality. Optimal use of these tools is within a comprehensive quality program.
Tool
Description
a
Cause-and-effect diagram
Check sheet
Control chartb
Histogram
Pareto chart
Scatter diagram
Stratificationc
TABLE II. Seven quality tools for management and planning. These tools are used for abstract analysis as well
as detailed planning.
Tool
Description
Affinity diagram
Relations diagram
Tree diagram
Matrix diagram
Matrix data analysisa
Arrow diagram
Process decision
program chart
a
1531
1531
consistent and up-to-date set of specifications for our procedures and equipment. Task Group reports that contain specifications e.g., TG-40,32 TG-53,33 etc. can be several years
behind technology implementation and not easily updated.
Furthermore, independent checks of subsystems are becoming more difficult as newer technologies, such as Tomotherapy HiArt and Accuracy CyberKnife, are self-contained
planning and delivery systems. Specifications are needed for
these new systems as well.
Error reporting mechanisms e.g., the ROSIS database,
www.clin.radfys.lu.se need continual development to allow
optimal sharing of information, which will help to understand clinical processes, equipment, and their failure modes.
Quality and error reduction should go hand-in-hand. When a
process is predictable, one can consider an error as being
built into the process, i.e., a given number of errors are guaranteed to occur. If high-quality processes with minimal
variation are developed and implemented, then fewer errors
should result. Variation, for example in a repeated set of
measurements, is an easy concept to appreciate. What may
not be so obvious is that clinical processes also contain
variation. Without an appreciation of variation, it is difficult
to predict the future of process operation or understand past
performance. It is also easy to blame others for errors over
which they have little control.
There are, however, situations where even a single error
may result in catastrophic loss, for example a miscalibration
of a new linear accelerator during acceptance or delivery
errors in a single-fraction radiosurgery treatment. A different
set of modern quality and error reduction tools are required
in these cases.34 Tools that can be useful are repeatability and
reproducibility studies, checklists, mistake-proofing, and the
usual independent verification checks.
High-quality patient service is a necessity. A goal should
be to understand and also believe what patients want and use
that as a compass to define optimal quality. Quality and error
reduction programs should consist of process design, process
implementation with statistical evaluation, and design improvement based on the data the process provides. Vague
statements on quality should no longer be acceptable such as
I think we are doing OK or things seem to be getting
better. The new requirement should be the evaluation and
documentation of process behavior based on the data.
IV. HOW DO WE GET THERE?
A modern approach to quality as found in most industries
must be implemented in radiation oncology. Modern quality
techniques developed in industry have already been suggested as important to healthcare.35,36 The concept of measurement and an appreciation of a system are essential to
achieve optimal quality. Measures of improvement should be
created together with a focus on specific projects that are
critical to a departments success. Quality should be restricted to problems of loss caused by variability of function
and related harmful side effects, or it will slip out of the
domain of medical physics into the psychological domain of
cultural or personal values.37 Facets of a modern approach to
1532
quality include senior management leadership, employee involvement and empowerment, patient defined quality patient satisfaction, a view of work as a system consisting of
different processes, and continuous improvement. Such models should help with gathering information and taking action
efficiently and effectively. In-depth education on quality is
needed along with skills transfer that will give a team and
individuals the power to lead sustainable change. Relevant
projects must be incorporated with broad training that is tied
to an overall vision. Picking out a single quality tool or technique will lead to unimpressive results and ultimately not
work. We believe the following six objectives are necessary
to achieve the goal of optimal quality.
A. Encourage research on quality
1532
Specification of operating limits for equipment and procedures is an essential part of developing a quality program. A
consistent, up-to-date specification document for all aspects
of radiation oncology is required. At the rate of new technology development and implementation, specifications will be
changing rapidly. The first step is to collate existing specifications e.g., Table II of TG-40 into one place. These documents should be condensed to the minimum necessary specifications rather than an exhaustive list of all possible things
to check. The specifications should be contained in a single
document or Web pages for all aspects of radiation oncology. This document should be live and readily available to
departments and vendors. A system of regular updates can be
accomplished by a joint organization with appointments
from the AAPM and ASTRO that has the authority to make
necessary changes. The organization or committee will discuss specifications for new equipment and procedures or revisit antiquated specifications. For brand new equipment, the
committee can recommend temporary specifications based
on the experience and input from early adopters. A start in
this direction is the AAPM Therapy Committees assignment
of a Quality Assurance and Outcome Improvement Subcommittee that is working to fast track recommendations for new
devices in the form of short Task Group reports. A subsequent initiative, after considerable research has been done, is
for the AAPM Task Groups to make recommendations on
organization- or department-wide quality programs and the
1533
use of modern quality tools. This step has begun with the
formation of TG-100, which has taken the approach to develop a framework for designing quality management activities. Specifically, TG-100 will investigate and present the
technique of Failure Modes and Effects Analysis with specific examples to IMRT and HDR brachytherapy. Lastly,
clear guidelines are needed for what constitutes quality, what
constitutes an error, and how to report errors. Other fields
have also been plagued with confusing nomenclature.51
D. Close collaboration with vendors
1533
The highest bar one can set is to treat patients as customers. It is easy for physicians and physicists to discount the
patients expectations of health quality. The science of quality is making its way into healthcare and patients may be the
greatest resource to determine what needs to be improved.53
Patient requirements should be related to quality characteristics of the service being provided. A way to achieve this is to
have patient advocates on departmental quality committees
as well as on AAPM and ASTRO quality committees. Quality characteristics identified with patient input should be targeted for improvement and translated into necessary functions of staff and equipment. Patient survey data will be an
important tool to learn about the patients perception of care.
But surveys must be used carefully as the potential for error
and bias can be significant.30 Quality improvement capacity
needs to be aligned with professional receptiveness, leadership, technical expertise, and survey data.54 It is important to
remember that the patient is the greatest beneficiary of an
optimal quality program.
V. CONCLUSION
Medical physicists and radiation oncologists have readily
harnessed new technologies and have made many significant
contributions to radiation oncology over the years. Much like
medicine, quality is also an art and there is a need for investigation. Scientific training leads physicists and oncologists
to wait for convincing evidence for the effectiveness of new
techniques before incorporating any change of procedures to
treat patients. However, our field should not quickly dismiss
approaches to quality that have roots in probability and statistics with many years of practical experience and demonstrated benefit in other fields.
ACKNOWLEDGMENTS
The authors would like to thank the Associate Editor and
referees for many constructive comments that made their
way into the final version of this manuscript.
a
1534
11
C. A. Perez, P. Gardner, and G. P. Glasgow, Radiotherapy quality assurance in clinical trials, Int. J. Radiat. Oncol., Biol., Phys. 10Suppl 1,
119125 1984.
12
R. M. Macklis, T. Meier, and M. S. Weinhous, Error rates in clinical
radiotherapy, J. Clin. Oncol. 16, 551556 1998.
13
G. Huang, G. Medlam, J. Lee, S. Billingsley, J. P. Bissonnette, J. Ringash,
G. Kane, and D. C. Hodgson, Error in the delivery of radiation therapy:
Results of a quality assurance review, Int. J. Radiat. Oncol., Biol., Phys.
61, 15901595 2005.
14
C. Szerlag, Best practices, ADVANCE 16, 8085 2006.
15
A. L. Boyer and T. Schultheiss, Effects of dosimetric and clinical uncertainty on complication-free local tumor control, Radiother. Oncol. 11,
6571 1988.
16
J. W. H. Leer, R. Corver, J. J. A. M Kraus, J. Ch. Togt, and O. J. S.
Buruma, A quality assurance system based on ISO standards: experience
in a radiotherapy department, Radiother. Oncol. 35, 7581 1995.
17
K. Holli, P. Laippala, A. Ojala, and M. Pitkanen, Quality control in
health care: an experiment in radiotherapy planning for breast cancer
patients after mastectomy, Int. J. Radiat. Oncol., Biol., Phys. 44, 827
833 1999.
18
M. A. Pitkanen, K. A. Holli, A. T. Ojala, and P. Laippala, Quality assurance in radiotherapy of breast cancervariability in planning target volume delineation, Acta Oncol. 40, 5055 2001.
19
B. Tomadsen, S. W. Lin, P. Laemmrich, T. Waller, A. Cheng, B. Caldwell,
R. Rankin, and J. Stitt, Analysis of treatment delivery errors in brachytherapy using formal risk analysis techniques, Int. J. Radiat. Oncol.,
Biol., Phys. 57, 14921508 2003.
20
T. Pawlicki, M. Whitaker, and A. L. Boyer, Statistical process control
for radiotherapy quality assurance, Med. Phys. 32, 27772786 2005.
21
G. A. Patton, D. K. Gaffney, and J. H. Moeller, Facilitation of radiotherapeutic error by computerized record and verify systems, Int. J. Radiat. Oncol., Biol., Phys. 56, 5057 2003.
22
W. E. Deming, The New Economics for Industry Government, Education,
2nd ed. MIT, Cambridge, MA, 1993.
23
P. Scalliet, Risk, society and system failure, Radiother. Oncol. 80, 275
281 2006.
24
A. Niemierko, Quality assurance and validation of individual IMRT
treatment plans before delivery, Med. Phys. 31, 421422 2004.
25
G. S. Ibbott, A. Molineu, and D. S. Followill, Independent evaluations of
IMRT through the use of an anthropomorphic phantom, Technol. Cancer
Res. Treat. 5, 481487 2006.
26
Abt Associates Inc., Washington DC, 2003, pp. 174.
27
ISO 9000 and Other Standards, American Society for Quality, http://
www.asq.org/learn-about-quality/iso-9000/overview/overview.html,
8
January 2007.
28
R. Lupan, I. C. Bacivarof, A. Kobi, and C. Robledo, A relationship
between Six Sigma and ISO 9000:2000, Qual. Eng. 17, 719725 2005.
29
M. R. OLeary, Lexikon, Joint Commission on Accreditation of Healthcare Organization JACHO, Oakbrook Terrace, 1994.
30
R. G. Carey and R. C. Lloyd, Measuring Quality Improvement in Healthcare: A Guide to Statistical Process Control Applications ASQ Quality,
Milwaukee, WI, 2001.
31
W. H. Ettinger, Six Sigma: adapting GEs lessons to health care,
Trustee 54, 1015 2001.
32
G. J. Kutcher, L. Coia, M. Gillin, W. F. Hanson, S. Leibel, R. J. Morton,
J. R. Palta, J. A. Purdy, L. E. Reinstein, and G. K. Svensson, Comprehensive QA for radiation oncology: report of AAPM Radiation Therapy
Committee Task Group 40, Med. Phys. 21, 581618 1994.
33
B. Fraass, K. Doppke, M. Hunt, G. Kutcher, G. Starkschall, R. Stern, and
J. Van Dyke, American Association of Physicists in Medicine Radiation
Therapy Committee Task Group 53: Quality assurance for clinical radio-
1534
therapy treatment planning, Med. Phys. 25, 17731829 1998.
D. Hutchison, Chaos theory, complexity theory, and health care quality
management, Qual. Prog. November, 6972 1994.
35
P. P. Harteloh and F. W. Verheggen, Quality assurance in health care.
From a traditional towards a modern approach, Health Policy 27, 261
270 1994.
36
G. Laffel and D. Blumenthal, The case for using industrial quality management science in health care organizations, JAMA, J. Am. Med. Assoc. 262, 28692873 1989.
37
G. Taguchi, Introduction to Quality Engineering American Supplier Institute, Dearborn, MI, 1986.
38
O. Hupe and J. Brunzendorf, A novel method of radiochromic film dosimetry using a color scanner, Med. Phys. 33, 40854094 2006.
39
A. L. McNiven, M. Mulligan, T. Kron, and J. J. Battista, The response of
prototype plane-parallel ionization chambers in small megavoltage x-ray
fields, Med. Phys. 33, 39974004 2006.
40
A. S. Meigooni, S. B. Awan, and K. Dou, Feasibility of calibrating
elongated brachytherapy sources using a well-type ionization chamber,
Med. Phys. 33, 41844189 2006.
41
A. Piermattei, A. Fidanzio, G. Stimato, L. Azario, L. Grimaldi, G.
DOnofrio, S. Cilla, M. Balducci, M. A. Gambacorta, N. Di Napoli, and
N. Cellini, In vivo dosimetry by an aSi-based EPID, Med. Phys. 33,
44144422 2006.
42
C. Talamonti, M. Casati, and M. Bucciolini, Pretreatment verification of
IMRT absolute dose distributions using a commercial a-Si EPID, Med.
Phys. 33, 43674378 2006.
43
S. Yoo, G. Y. Kim, R. Hammoud, E. Elder, T. Pawlicki, H. Guan, T. Fox,
G. Luxton, F. F. Yin, and P. Munro, A quality assurance program for the
on-board imager, Med. Phys. 33, 44314447 2006.
44
O. A. Zeidan, S. A. L. Stephenson, S. L. Meeks, T. H. Wagner, T. R.
Willoughby, P. A. Kupelian, and K. M. Langen, Characterization and use
of EBT radiochromic film for IMRT dose verification, Med. Phys. 33,
40644072 2006.
45
D. Thwaites, Quality assurance into the next century, Radiother. Oncol.
54, viiix 2000.
46
J. E. Ibrahim, Translating quality into research: Do we need more research into quality or should quality activities be conducted using the
principles and methodological rigour of scientific research?, J. Qual.
Clin. Pract. 20, 6364 2000.
47
H. Singh, L. A. Petersen, and E. J. Thomas, Understanding diagnostic
errors in medicine: a lesson from aviation, Qual. Saf. Health. Care 15,
159164 2006.
48
P. Dixon and B. OSullivan, Radiotherapy quality assurance: time for
everyone to take it seriously, Eur. J. Cancer 39, 423429 2003.
49
P. B. Boggs, Peak expiratory flow rate control chart: a breakthrough in
asthma care, Ann. Allergy Asthma Immunol. 77, 429432 1996.
50
T. Kehoe and L.-J. Rugg, From technical quality assurance of radiotherapy to a comprehensive quality of service management system, Radiother. Oncol. 51, 281290 1999.
51
K. H. Yu, R. L. Nation, and M. J. Dooley, Multiplicity of medication
safety terms, definitions and functional meanings: when is enough
enough?, Qual. Saf. Health. Care 14, 358363 2005.
52
P. Pronovost and R. Wachter, Proposed standard for quality improvement research and publication: one step forward and two steps back,
Qual. Saf. Health. Care 15, 152153 2006.
53
B. L. Sadler, To the class of 2005: Will you be ready for the quality
revolution?, J. Qual. Pat. Saf. 32, 5455 2006.
54
E. Davies and P. D. Cleary, Hearing the patients voice? Factors affecting the use of patient survey data in quality improvement, Qual. Saf.
Health. Care 14, 428432 2005.
34
Received 26 February 2007; revised 2 May 2007; accepted for publication 2 May 2007;
published 11 June 2007
How will the future of picture archiving and communication systems PACS look, and how will
this future affect the practice of radiology? We are currently experiencing disruptive innovations
that will force an architectural redesign, making the majority of todays commercial PACS obsolete
as the field matures and expands to include imaging throughout the medical enterprise. The common architecture used for PACS cannot handle the massive amounts of data being generated by
even current versions of computed tomography and magnetic resonance scanners. If a PACS cannot
handle todays technology, what will happen as the field expands to encompass pathology imaging,
cone-beam reconstruction, and multispectral imaging? The ability of these new technologies to
enhance research and clinical care will be impaired if PACS architectures are not prepared to
support them. In attempting a structured approach to predictions about the future of PACS, we offer
projections about the technologies underlying PACS as well as the evolution of standards development and the changing needs of a broad range of medical imaging. Simplified models of the history
of the PACS industry are mined for the assumptions they provide about future innovations and
trends. The physicist frequently participates in or directs technical assessments for medical equipment, and many physicists have extended these activities to include imaging informatics. It is hoped
that by applying these speculative but experienced-based predictions, the interested medical physicist will be better able to take the lead in setting information technology strategies that will help
facilities not only prepare for the future but continue to enjoy the benefits of technological innovations without disruptive, expensive, and unexpected changes in architecture. A good PACS strategy can help accelerate the time required for innovations to go from the drawing board to clinical
implementation. 2007 American Association of Physicists in Medicine.
DOI: 10.1118/1.2743097
Key words: PACS, information systems, Moores law, technology assessment
I. INTRODUCTION
Why make predictions about the future of picture archiving
and communication systems PACS? Our ability to deliver
quality care is increasingly dependent upon information technologies IT that drive all aspects of image acquisition,
communication, interpretation, and storage. A radiology department can no longer operate with film and expect to effectively process and read a computed tomography CT
study from a modern scanner. Architectural decisions about
todays PACS technology can lead to limitations where new
imaging modalities and techniques are not supported or image data cannot be economically stored and retrieved adequately for medical management decisions. The ability to
grow and innovate relies on having an, open, flexible, and
scalable architecture. This paper presents six predictions that,
if taken together, present a scenario for the next generation of
PACS.
We are taught as scientists that all extrapolations including, perhaps especially, predictions are fraught with uncertainty, so that it is important to clearly state the assumptions
on which such extrapolations are based. One of the key assumptions in looking to the future of PACS is that the industry has a relatively small market size compared with other IT
industries. Despite its mission-critical clinical application,
2676
this market is still being driven by medical device manufacturers. Those factors are important in the PACS industrys
somewhat later adopter path in the classic pattern of technologic innovations when compared with other computer
industry sectors. By looking at other domains in the computer industry, therefore, we can make modest predictions
about the future of PACS with some confidence. Noted futurist William Gibson is often quoted as saying, The future
is here. Its just not widely distributed yet.1 These predictions, too, may not occur at precisely the forecast intervals
but, barring extraordinary intervening circumstances, will almost certainly prove valid.
Prediction 1. Moores law will hold true for at least
another 10 years
0094-2405/2007/347/2676/7/$23.00
2676
2677
2677
2678
2678
FIG. 2. Storage usage per year has seen growth predominantly from an
escalation in the number of CT studies and CT study volume.
2679
2679
2680
Volumetric imaging has brought both progress and advantages to visualization and parameterization of organ systems
and pathologies in the past 10 years, thanks in no small part
to the medical physics community. At the same time, the
range of visualization techniques has expanded rapidly along
with new modalities, hybrid imaging technologies, and innovative procedures. No single PACS vendor can innovate in
all the subspeciality areas of image visualization and analysis, including but not limited to mammography, ultrasound,
nuclear medicine, cardiology, chest imaging, perfusion imaging, functional imaging, registration and fusion imaging, CT
colonoscopy, not to mention all the new tomosynthesis and
CAD applications coming to market.
One disturbing byproduct of these advances has been the
increasing proliferation of specialty workstations with limited integration into clinical workflow. It is becoming inMedical Physics, Vol. 34, No. 7, July 2007
2680
2681
emphasis will become increasingly important on open standard integration. Although the DICOM transfer syntax might
evolve, the main DICOM object model will be relatively
unchanged in 10 years and will be one of the few things
recognizable to systems used today.
II. CONCLUSION
Institutions should develop strategies for future proofing their technology buying decisions. The most important
factor is to ensure that the purchasing model allows the institution to enjoy the market benefits of Moores law. The
most direct way to accomplish this is to move to a softwareonly model in which hardware purchases are made independently of the PACS vendor, with only minimum hardware
requirements for servers, storage, and workstations. This allows a site to regularly replace hardware at market value and
enjoy benefits in better performance, reliability, and cost.
Another recommended task is to hold discussions with the
PACS vendor on ways to incorporate third-party workstations into the clinical workflow. The lowest cost, long-term
solution for customers is integration through openly published standards and not piecemeal vendor partnerships with
proprietary interfaces. Vendors should also provide a roadmap detailing the ways in which they plan to incorporate
volumetric datasets for visualization and archiving without
compromising the productivity of the radiologists.
Physicists have a real opportunity to provide the technical
leadership that will help to shape strategies as radiology departments meet the challenges of the future. Physicists are
ideally suited to provide informatics leadership because of
our proficiency with technology, intimate domain knowledge
of imaging, and close partnership with clinical staff and physicians. Informatics is a natural extension for many physicists and an avenue for both career growth and contributions
to operations and productivity. Important discussions in the
medical physics community have focused on the role of the
physicist in implementing and supporting information systems in a hospital.28,29 Both sides agree that IT is certainly an
opportunity; the debate is over whether changes should be
made to the requirements of an already large curriculum of
physicist training. It is clear that more discussion is necessary on the new and challenging opportunities in the practice
of medical physics. What is undisputed is that unique to the
medical discipline, the radiologist and the physicist have had
a mutually supportive relationship for more than a century.30
The radiologist needs IT guidance and support in information
systems now more than ever before.
ACKNOWLEDGMENTS
I would like to thank Dr. Nancy Knight from the University of Maryland School of Medicine and Dr. William
Hendee from the Medical College of Wisconsin for their expert assistance in preparing this manuscript.
1
2681
15 December 2006.
G. Moore, Cramming more components onto integrated circuits, Electronics
38, 114117 1965; ftp://download.intel.com/museum/
Moores_Law/Articles-Press_Releases/Gordon_Moore_1965_Article.pdf,
8 January 2007.
4
V. Freeman, Athlon 64 X2 4800+ processor review, Sharkey Extreme,
June 22, 2005. http://www.sharkyextreme.com/hardware/cpu/article.php/
3514901, 9 January 2007.
5
S. Levy, The hard disk that changed the world, Newsweek, August 7,
2006, http://www.msnbc.msn.com/id/14096484/site/newsweek/, 8 January 2007.
6
A brief history of the hard disk drive, DUX Computer Digest, May 19,
2005. http://www.duxcw.com/digest/guides/hd/hd2.htm, 8 January 2007.
7
S. Olsen, Hitachi Global Storage announces 1TB Hard Drive, Daily
Tech., January 5, 2007. http://www.dailytech.com/article.aspx?newsid
5590, 8 January 2007.
8
M. Kanellos, Intel scientists find wall for Moores Law, ZDNet News,
December 1, 2003. http://news.zdnet.com/2100-9584_22_5112061.html,
9 January 2007.
9
D. Mohney, Moores Law meets market saturation, The Inquirer, December 5, 2002. http://www.theinquirer.net/default.aspx?article6559, 9
January 2007.
10
Rocks law of the doubling of fabrication costs, Wikipedia, the Free
Encyclopedia, http://en.wikipedia.org/wiki/Rock%27s_law, 15 December
2006.
11
M. Kanellos, Moores Law to roll on for another decade, CNET News,
February 10, 2003, http://news.com.com/2100-1001-984051.html, 9 January 2007.
12
B. Schaller, The origin, nature, and implications of Moores Law, Microsoft Research, September 26, 1996; http://research.microsoft.com/
~gray/Moore_Law.html, 9 January 2007.
13
P. Gargini, Sailing with the ITRS into nanotechnology, PowerPoint
presentation,
ftp://download.intel.com/technology/silicon/
Paolo_Semicon_West_071904.pdf, 15 December 2006.
14
C. M. Christenson, The Innovators Dilema: The Revolutionary Book that
Will Change the Way You Do Business Collins Business Essentials, New
York, 2003.
15
K. Schwartz, Nexan gives ATA more muscle with SATABeast, eWeek.com,
October
18,
2005,
http://www.eweek.com/article2/
0,1895,1872607,00.asp, 9 January 2007.
16
B. J. Erickson, K. R. Persons, N. J. Hangiandreou, E. M. James, C. J.
Hanna, and D. G. Gehring, Requirements for an enterprise digital image
archive, J. Digit Imaging 14, 7282 2001.
17
C. Meenan, B. Daly, C. Toland, and P. Nagy, Use of a thin-section
archive and enterprise 3D software for long-term storage of thin-slice CT
datasets. J. Digit Imaging 19suppl 1, 8488 2006.
18
F. M. Behlen, A DICOM document-oriented approach to PACS infrastructure, J. Digit Imaging 113 suppl. 1, 3538 1998.
19
F. M. Behlen, R. E. Sayre, J. B. Weldy, and J. S. Michael, Permanent
records: Experience with data migration in radiology information system
and picture archiving and communication system replacement, J. Digit
Imaging 13 2 suppl. 1, 171174 2000.
20
P. Thibodeau, Flash memory not ready to explode as PC storage technology,
Computer
World,
April
3,
2007,
http://
www.computerworld.com/action/article.do?command
viewArticleBasic&articleId9015478, 15 April 2007.
21
M. Fontecchio, Blade server growth drives revenue Q3 06, SearchDataCenter.com,
November
22,
2006.
http//
searchdatacenter.techtarget.com/originalContent/
0,289142,sid80_gci1231203,00.html, 9 January, 2007.
22
Hewlett-Packard, Inc, HP BladeSystem c-class servers blades
interconnects, News Release, http://h18004.www1.hp.com/products/
blades/components/c-class-interconnects.html, 9 January 2007.
23
DM Review Editorial Staff, The 24 7 challenge: IOUG 2006 survey on
high availability trends, DM Review, August 29, 2006, http://
www.dmreview.com/editorial/newsletter_article.cfm?articleId1061768,
9 January 2007.
24
Google, Wikipedia, the Free Encyclopedia, http://en.wikipedia.org/
wiki/Google#Technology, 31 January 2007.
25
T. Wilson and J. Tyson, How graphics cards work, HowStuffWorks,
http://computer.howstuffworks.com/graphics-card.htm, 9 January 2007.
26
N. D. Terry, Introduction to OpenGL, Tulane University Class CPEN
3
2682
2682
29
X. Allen Lia
Department of Radiation Oncology, Medical College of Wisconsin, 8701 Watertown Plank Road,
Milwaukee, Wisconsin 53226
Received 3 January 2007; revised 9 August 2007; accepted for publication 10 August 2007;
published 11 September 2007
Rapid advances in functional and biological imaging, predictive assays, and our understanding of
the molecular and cellular responses underpinning treatment outcomes herald the coming of the
long-sought goal of implementing patient-specific biologically guided radiation therapy BGRT in
the clinic. Biological imaging and predictive assays have the potential to provide patient-specific,
three-dimensional information to characterize the radiation response characteristics of tumor and
normal structures. Within the next decade, it will be possible to combine such information with
advanced delivery technologies to design and deliver biologically conformed, individualized therapies in the clinic. The full implementation of BGRT in the clinic will require new technologies and
additional research. However, even the partial implementation of BGRT treatment planning may
have the potential to substantially impact clinical outcomes. 2007 American Association of
Physicists in Medicine. DOI: 10.1118/1.2779861
Key words: biologically guided radiation therapy, biological imaging, outcome modeling, LQ
model
I. INTRODUCTION
The efficacy of radiotherapy RT is ultimately determined
by our ability to deliver doses of radiation sufficient to eradicate a tumor, while adequately sparing nearby normal structures. Until now, the practice of RT has been based on the
premise that effective treatments require the delivery of uniform radiation doses to all target volumes. The uniform-dose
theorem1 provides some justification for this treatment strategy. However, the uniform-dose approach only yields the
best possible tumor control for the implausible situation in
which all regions of the tumor have exactly the same biological characteristics and sensitivities to radiation. Biological factors that modulate treatment outcomes include such
factors and processes as a cells capacity for repair, acute and
chronic hypoxia, cell proliferation, the spatial distribution
and density of tumor clonogens, and the differential sensitivity of cells to radiation during the cell cycle. Genetic and
other patient-specific factors that impact on radiation responses are seldom considered in radiation treatment planning for specific patients.
Theoretical studies and the accumulated clinical data214
strongly suggest that the treatment plans designed to exploit
patient- and tumor-specific biological features will substantially improve treatment outcomes. However, the complexity
of biological systems and our incomplete knowledge of the
mechanisms underpinning tumor and normal-tissue responses, pose many challenges for the clinical implementation of biological criteria into the treatment planning process.
To advance patient-specific treatment planning without unduly jeopardizing patient care, we propose a three-phase
3739
0094-2405/2007/3410/3739/13/$23.00
3739
3740
t=
ex
2dx
Deff TD50
mTD50
Deff =
,
viD1/n
i
3740
i=1
3741
3741
3742
SD = exp D 1 +
GD
/
2
D2
t
dtD
tett ,
dtD
2 x
G1
=
e + x 1,
n nx2
is more appropriate. Here, x ln2T1 / and T1 is the fraction delivery time time to deliver one fraction. The time to
Medical Physics, Vol. 34, No. 10, October 2007
3742
deliver a fraction in IMRT has the potential to have a significant impact on tumor control for tumors with a low / ratio
and a short repair half-time.73
Although the mechanistic basis of the LQ has been challenged and remains open to vigorous debate,7477 the interpretation of the LQ survival model in terms of DSB induction and processing provides an explanation for the linearquadratic dependence on dose of the surviving fraction and,
equally important, provides a conceptual framework to understand the interplay between damage repair processes and
dose protraction dose rate effects. Studies showing that defects in DSB repair mechanisms reduce cell survival directly
implicate DSB processing in survival responses.78,79 In addition, positive correlations among chromosomal damage and
clonogenic survival provide strong circumstantial evidence
that DSB induction and processing is an important cell killing mechanism. In one study, Cornforth and Bedford80 demonstrated that, in plateau-phase cultures of AG1522 normal
human fibroblasts, asymmetric exchanges exhibited a near
one-to-one correlation with the logarithm of the surviving
fraction, as would be predicted by Eq. 5 when D
+ GD2 is interpreted as the expected number of lethal aberrations per cell. Studies such as this one suggest that the
conversion of DSB into lethal exchanges through intra- and
intertrack binary misrepair are an especially important cellkilling mechanism and provides some justification for considering the LQ as more than a purely phenomenological
model.
The mathematical simplicity of the LQ survival model is
both its strength and weakness. Low-dose hyperradiosensitivity and adaptive responses notwithstanding,8183
the strength of the LQ is that trends in clonogenic survival
can often be adequately predicted in vitro and in vivo using a
minimum number of adjustable parameters , / , and or
. However, when contrasted to the underlying biology, the
LQ model can only be viewed as a highly idealized representation of a very complex, time-dependent sequence of
biophysical events and processes. That is, the aggregate effects of many time- and cell-cycle dependent processes are
represented by a single set of time- and cell-cycle independent radiosensitivity parameters, , / , and or . This
oversimplification of the underlying biology implies that the
collective radiation response of a population of cells, even if
this cell population has uniform and stable genetic traits, is
determined by a distribution of values for , / , and
rather than one true set of values. This inherent variability
in cellular radiosensitivity is further amplified by a host of
other patient- or tumor-specific factors.8486
Our ability to accurately estimate a TCP using the LQ
model is ultimately limited by our lack of knowledge of the
most appropriate parameter distributions for , / , and
and by our incomplete knowledge of how well the LQ model
relates dose to clonogenic survival reproductive death. To
reduce the possibility that treatment plans are inappropriately
overoptimized for a specific combination of radiosensitivity
parameters, distributions of values for , / , and should
be used to predict treatment indicators e.g., EUD or BED
and outcomes rather than point estimates.8790 The use of
3743
3743
FIG. 1. Prescription doses for the treatment of prostate cancer equivalent to 37 daily fractions of 2 Gy filled circle. A fraction delivery time T1 parameter
of 10 min is used to compute G1 for all treatments refer to Eq. 7. Solid lines denote prescription doses estimated using the Fowler et al. Ref. 91
radiosensitivity parameters. Dashed lines indicate prescription doses estimated using the Wang et al. Ref. 92 radiosensitivity parameters.
Although uncertainties in radiosensitivity parameters appear an overwhelming obstacle for the direct application of
biological models in treatment planning, many issues associated with the inadequacy of biological models and radiosensitivity parameters can be circumvented through the use of
isoeffect calculations. Consider as an example the task of
identifying biologically equivalent prescription doses for the
treatment of prostate cancer. To identify isoeffective total
treatment doses, correct Eq. 5 for repopulation effects and
then take the logarithm and apply the quadratic formula to
obtain
D=
/
1+
2G
/
1+
2G
4Gln S T
/
1+
4G
T
BED +
/
3744
3744
FIG. 2. Potential impact of interpatient heterogeneity on the determination of isoeffective prescription doses. All prescription doses are equivalent to a
reference treatment composed of 37 fractions of 2 Gy filled circles. A fraction delivery time T1 parameter of 10 min is used to compute G1 for all
treatments refer to Eq. 7. The solid and dotted lines are the mean and 95% confidence interval, respectively, derived from estimated prescription doses for
104 patients. Patient radiosensitivity parameters sampled from a uniform probability density function using Monte Carlo methods: 0.1 Gy1 to 0.3 Gy1,
/ 1 Gy to 10 Gy, 0.1 h to 6 h, and Td 30 d to 365 d.
the form of a clinically accepted treatment, is explicitly considered e.g., the filled circles in Figs. 1 and 2. In addition,
Eq. 8 suggests an easy way to manage the inherent risks
associated with altering an accepted treatment, i.e., manage
risk by making small rather than large changes in the number
of daily fractions. The results shown in Figs. 1 and 2 suggest
that a change from 37 fractions of 2 Gy to n = 30 or to n
= 50 may be accomplished with a nominal risk for altered
tumor control. Large changes in an accepted treatment e.g.,
n = 37 to n = 5 or 10 are potentially risky because of interpatient heterogeneity. Large changes in an accepted treatment
are also risky because altering the fractionation schedule may
substantially alter normal tissue toxicity. Small changes in an
accepted treatment plan are also advisable to minimize the
potential for unanticipated consequences associated with the
approximations and simplifications inherent in the LQ model
and other biological factors, e.g., bystander effects and lowdose hyper-radiosensitivity.
In addition to interpatient heterogeneity, radiosensitivity
parameters vary within a tumor, because, for example, of
genomic instability and developmental or functional disturbances in tumor vasculature. Developmental and functional
disturbances in the tumor vasculature produce varying levels
of acute and chronic hypoxia93 and are often associated with
more advanced stages of disease.94,95 Cells irradiated under
hypoxic conditions are much less sensitive to radiation, and
substantially higher doses may be necessary to inactivate tumor cells in regions of a tumor with intermediate or extreme
levels of hypoxia. Intratumor variations in radiosensitivity
parameters enhance the risks associated with making large
changes in clinically accepted treatment plans. However, in-
3745
tratumor heterogeneity is also a potential opportunity to improve treatment outcomes through techniques such as dose
painting by numbers.9698 The objective of dose painting by
numbers is to use biological information to identify a dose
distribution that achieves biological rather than physical
dose conformity.
The attempt to identify a biologically conformal dose distribution can be cast in terms of isoeffect calculations. To
illustrate, imagine that a technique is developed to noninvasively estimate oxygen pO2 levels within a tumor. The
measured pO2 level can then be converted to an oxygen enhancement ratio OER using the formula99 OERi = mK
+ O2i / K + O2i, where m = 2.7, K = 0.25, and O2i denotes the oxygen concentration in the ith region of the tumor.
This OER may be interpreted as the ratio of the dose to the
hypoxic cells to the dose to the aerobic cells required to
produce the same number of DSB per cell,99 and it ranges
from one for fully aerobic cells to about three for extreme
levels of hypoxia. The analyses of Carlson et al.99 suggest
that OER values are nearly independent of cellular radiation
sensitivity i.e., and / .
For cells irradiated in vitro, Carlson et al.99 have shown
that radiosensitivity parameters for hypoxic H and aerobic
A cells are well approximated by H = A / OER and
/ H = OER / A. The relationships among radiosensitivity parameters for aerobic and hypoxic cells imply that the
dose to regions of the tumor with reduced oxygen levels
needs to be increased by a factor equal to the OER to achieve
the same surviving fraction as well oxygenated regions of the
tumor.99,100 Hence, a biologically conformal dose distribution
might be achieved by designing a treatment to deliver dose
Di = OERi D to the ith region of the tumor. Here, D is a
clinically accepted prescription dose.
The suggested approach to individualize a treatment to
correct for the effects of hypoxia will necessitate a validated
technique to determine pO2 levels within the tumor. In addition, the application of the suggested strategy is not without
risk because of uncertainties associated with the formula to
determine the OER. However, the observation that isoeffect
calculations are relatively insensitive to uncertainties in biological parameters e.g., Figs. 1 and 2 suggests that the approach may still provide useful guidance for treatment individualization. The example also illustrates how future
technological developments and knowledge of the molecular
and cellular mechanisms underlying radiation sensitivity
might be translated into the clinic and used to guide the
treatment planning process.
II.F. Research needs for outcome modeling
Outcome modeling is challenging in part because biological parameters used in these models are patient, tumor, and
organ specific101,102 and in part because TCP and NTCP models are highly nonlinear. That is, seemingly small, patientspecific differences in physical or biological factors can result in very different clinical outcomes for treatment plans
that are otherwise identical. As illustrated by the very different radiosensitivity parameters reported by Fowler et al.91
Medical Physics, Vol. 34, No. 10, October 2007
3745
3746
3746
Technologies for RT delivery, such as IMRT, imageguided IMRT IG-IMRT, novel brachytherapy, targeted RT,
and proton and heavy ion therapy, have rapidly advanced
over the last ten years, and this trend will most likely continue for the next decade. Advances in RT delivery, particularly in IG-IMRT, will enable the direct integration of not
only anatomic, but also biological images into treatment
units to guide the delivery process. The 4D space+ time
anatomic and biological imaging using CT, MRI, PET and
other modalities will enable the treatment of moving targets
and substantially reduce or eliminate the inadvertent irradiation of normal tissues. Techniques for charged particle
e.g., protons, helium, and carbon ions therapy will advance
and provide new capabilities in the clinic for delivery of
biologically conformed RT.
Although the physical and biological methods and technologies needed to implement BGRT are advancing quickly
and hold great promise for the future of RT, the potential
costs of implementing some technologies may be found prohibitive for routine use in the clinic. The integration of the
multiple technologies required for BGRT will require extensive clinical validation. In particular, the use of incomplete or
inaccurate biological information to preferentially target selected regions of a tumor at the expense of other tumor regions may jeopardize treatment outcomes. For reasons such
as these, we believe an incremental approach is advisable for
the clinical implementation of BGRT, as illustrated in Fig. 4
and briefly explained below.
3747
3747
3748
IV. SUMMARY
The ongoing and rapid development of RT technology,
such as IMRT, IG-IMRT, novel brachytherapy, targeted RT,
proton and heavy ion therapies, technologies for the characterization of the human genome, and the dazzling advances
in biological and functional imaging modalities have significantly increased the potential usefulness of implementing
BGRT treatment planning in the clinic. Biological image modalities, which are currently being introduced into the clinic,
are expected to improve the definition of tumor targets and
normal structures and provide patient-specific information
that can be used to guide the treatment planning. New imaging modalities will also provide new opportunities to quantify tumor and normal-tissue responses during or after a
course of radiation therapy. The ability to rapidly assess
treatment outcomes with surrogates for long-term diseasefree survival will in turn speed our ability to extract useful
information about the biological effectiveness of alternate
and refined radiation treatments.
The integration of biological imaging, radiation response
relationships, and knowledge of the molecular basis of cancer has the potential to dramatically improve RT outcomes
by tailoring the treatment process to the biological characteristics of specific patients. Combining advanced delivery
techniques, such as IG-IMRT, with the selection of patientspecific, biologically conformal prescription doses is the next
frontier in RT and substantial progress is likely within the
coming decade. We believe the three-phase strategy to implement BGRT in the clinic outlined in this paper has the potential to substantially advance patient-specific treatment
planning while minimizing the risks associated with the introduction of new technologies for patient care. The introMedical Physics, Vol. 34, No. 10, October 2007
3748
duction of patient-specific BGRT into the clinic can be accelerated through a multifaceted research program to
improve biological and functional imaging, predictive assays, and biophysical modeling of treatment outcomes. New
treatment planning tools that incorporate biological objectives into the treatment planning process are also needed.
The accumulation and analysis of clinical data for modern
treatment modalities, and the preclinical and clinical evaluation of new tools and techniques for patient-specific treatment planning is highly desirable.
Although the technologies and techniques needed for the
full implementation of patient-specific treatment planning
may require more than a decade to develop and validate,
even the partial implementation of BGRT treatment planning
has the potential to enhance local tumor control without an
undue increase in damage to normal tissues. The validation
and implementation of robust tools and technologies for the
prediction of patient-specific outcomes also has the potential
to improve the efficiency and costs of delivering RT. Why
deliver a treatment consisting of six to eight weeks of daily
fractions if an alternate plan with the same treatment outcome can be achieved with a plan, such as stereotactic RT or
accelerated hypofractionated RT, that only requires one or
two weeks to deliver? The coming decade will surely be an
exciting time for the practice of BGRT.
ACKNOWLEDGMENT
This work is supported in part by a research grant from
Siemens Oncology Care Systems.
a
3749
3749
3750
83
3750
3751
3751
therapy of gliomas: An application of 1H MRSI, J. Magn. Reson Imaging 16, 464476 2002.
113
W. R. Hendee, Biomedical imaging research opportunities workshop II:
A summary of findings and recommendations, Med. Phys. 32, 2484
2486 2005.
114
G. P. Chen, E. Ahunbay, C. J. Schultz, and X. A. Li, Development of an
inverse optimization package to plan non-uniform dose distributions
based on spatially inhomogeneous radiosensitivity extracted from biological images, Med. Phys. 34, 11981205 2007.
115
K. M. Schmainda, S. D. Rand, A. M. Joseph, R. Lund, B. D. Ward, A. P.
Pathak, J. L. Ulmer, M. A. Baddrudoja, and H. G. J. Krouwer, Characterization of a first-pass gradient-echo spin-echo method to predict brain
tumor grade and angiogenesis, AJNR Am. J. Neuroradiol. 25, 1524
1532 2004.
Received 6 July 2007; revised 30 August 2007; accepted for publication 31 August 2007;
published 15 October 2007
The interpretation of medical images by radiologists is primarily and fundamentally a subjective
activity, but there are a number of clinical applications such as tumor imaging where quantitative
imaging QI metrics such as tumor growth rate would be valuable to the patients care. It is
predicted that the subjective interpretive environment of the past will, over the next decade, evolve
toward the increased use of quantitative metrics for evaluating patient health from images. The
increasing sophistication and resolution of modern tomographic scanners promote the development
of meaningful quantitative end points, determined from images which are in turn produced using
well-controlled imaging protocols. For the QI environment to expand, medical physicists, physicians, other researchers and equipment vendors need to work collaboratively to develop the quantitative protocols for imaging, scanner calibrations, and robust analytical software that will lead to
the routine inclusion of quantitative parameters in the diagnosis and therapeutic assessment of
human health. Most importantly, quantitative metrics need to be developed which have genuine
impact on patient diagnosis and welfare, and only then will QI techniques become integrated into
the clinical environment. 2007 American Association of Physicists in Medicine.
DOI: 10.1118/1.2789501
Key words: quantitative imaging, cancer, computed tomography CT, positron emission tomography PET, single photon emission computed tomography SPECT, magnetic resonance imaging
MRI
I. CURRENT STATUS
Radiological diagnosis performed by radiologists in the current era is almost completely qualitative. Qualitative is not a
bad thingthe diagnostic utility of medical imaging, with
qualitative interpretation by an experienced radiologist, represents one of the greatest advances in modern medicine1 of
the 20th century. In the 1970s, when currently practicing
senior radiologists were trained, the practice of radiology
focused primarily on screen-film radiography. For example,
in 1975, computed tomography CT was fledgling, rectilinear scanners were only slowly being replaced by projection
nuclear scanners using Anger logic, B-mode ultrasound imaging was grainy and analog, and magnetic resonance imaging MRI was a crude sketch in Lauterburs laboratory
book. During this era, the hallmark of a radiologists talents
lay in his or her diagnostic skills in radiographic interpretation, fundamentally a qualitative experience. While some basic quantitative measures such as tumor width or anatomically relevant angles were and are still made on radiographs
using a ruler or protractor in scoliosis, for example, the fact
is that screen-film radiography does not lend itself naturally
to robust quantitative methods.
II. ROLE OF QUANTITATIVE IMAGING IN THE
FUTURE
The practice of radiology today has completely changed
relative to 20 years ago. Radiography, even digital radiography, is declining as a fraction of radiologists workload in the
United States, with tomographic modalities such as CT,
4173
MRI, ultrasound, SPECT, and PET becoming the predominant modalities used in imaging todays patients. The potential of quantitative imaging QI from tomographic data sets
is clearly superior to that of projection images. The modern
radiology department, in the academic or private practice
settings, is completely digital and PACS based. Digital tomographic volume data sets lend themselves more readily to
quantitative analysis than screen-film radiographs.
Radiology residents and fellows in training now have
spent considerable time during their youth with a joystick in
their hands playing video games, defending earth from the
Covenant, high jacking sports cars, or shooting up Liberty
City. This generation of radiologists is primed and ready to
do real time fly-bys of patients high resolution 3D anatomy.
Unfortunately, the performance of current $100,000 medical
PACS workstations in radiology departments pale in comparison to the graphical capabilities of modern high-end
video game hardware, which typically incorporate both parallel and pipeline processing techniques to accelerate graphic
visualization.
Modern imaging systems produce images which are rich
in quantitative information. While most tomographic modalities, including ultrasound imaging and MRI, possess solid
quantitative spatial information, CT arguably is at least today the modality which produces image data sets which are
the richest in quantitative anatomical information. The reasons for this are threefold: 1 CT in typical clinical practice
has the highest spatial resolution smallest voxel volume
with the MTF to back it up of tomographic modalities, mak-
0094-2405/2007/3411/4173/7/$23.00
4173
4174
4174
4175
4175
4176
Cancer patients with solid tumors undergoing chemotherapy are often subjected to serial CT studies with or without contrast injection to evaluate whether or not the size of
the lesion is changing over time. This type of study requires
that tumor size be compared at different time points during
the treatment. While some would argue that CT is a crude
metric to determine tumor viability compared to PET or
MRS or other imaging biomarkers, it is nevertheless the
most ubiquitous QI measure used today and therefore serves
as a good example.
IV.B. Acquisition protocol
4176
specific tumor type e.g., hepatocellular carcinoma to a specific drug, the time between the base line scan and a given
follow up scan should be controlled fixed to a specific value
with small leeway, such as 3 months plus or minus 1 week.
A CT protocol may scan the entire abdomen of the patient
using thicker CT sections at lower mA s values and therefore lower radiation levels for the same noise. For better
accuracy, the scan protocol could use thinner sections and
higher mA s near the anatomical site of known tumors to
produce lower noise, higher fidelity images. Contrast injection, if used, should be consistent in terms of type, volume
injected, injection rate, and delay between injection and imaging.
There are obviously a whole host of other parameters that
can and should be considered and set, because consistency is
needed between scans to achieve accurate quantitative results. Only a few acquisition parameters are discussed above,
in the interest of brevity.
IV.C. Image calibration
The RECIST criteria57 call for unidimensional measurements of a tumor at its widest point in-plane and there are a
set of other criteria required when multiple tumors are
present. Although the RECIST standard should be used short
term on NCI sponsored clinical trials, with the thin section
CT imaging capabilities of modern multislice CT scanners,
there is a strong sentiment in the QI community based on
simple reasoning that volume measurements are more accurate than linear measurements.
Most PACS workstations have basic measurement tools
such as distance and area, but these are often tedious and
time consuming to use. While there are many efforts underway to build easier and more automatic tumor segmentation
tools, getting these tools onto a given PACS system is not
always easy, and if these tools are to be used to affect patient
care, then only FDA approved software packages can be
4177
4177
It is incumbent upon software engineers and PACS vendors to provide hooks into the display software that comprises a PACS program which allow researchers and others
to add QI software packages to the standard PACS display
capabilities. Only if QI techniques can be bundled into an
intuitive interface on the PACS console will QI techniques
become adopted by radiologists. The software will need to be
robust and operate in a time-efficient manner. Obviously,
FDA approval for these packages is required for routine
clinical use, but hooks into PACS software for QI research is
an important start toward the long term adoption of quantitative measures in imaging.
Reporting quantitative metrics such as SUV or tumor volume in the free text portion of a radiologists report is inadequate and impractical. Radiology information system RIS
providers need to provide the ability to define special fields
in the radiologist report, where specific quantitative information can be located. Examples include tumor volume or tumor SUV value, for an array of tumors a patient might have.
These data fields need to be accessible by other databases, so
that tumor growth rate etc. can be automatically computed
for a given patient by the push of a button. For example, one
cancer patient could have five distinct tumors and four different CT scans over time. Hand computing 15 different volume changes from the free text reports is too time consuming
and is simply not feasible for routine care. This is what databases and computers are supposed to do, convert menial
labor to a push button procedure, but before this happens in
4178
4178
ume and area, Neuropsychology 21, 235241 2007.
F. P. Macmaster, R. El-Sheikh, A. R. Upadhyaya, J. Nutche, D. R. Rosenberg, and M. Keshavan, Effect of antipsychotics on pituitary gland volume in treatment-naive first-episode schizophrenia: A pilot study,
Schizophr. Res. 92, 207210 2007.
18
R. W. Katzberg, C. L. Dumoulin, M. A. Buonocore, L. R. Opsahl, R. D.
Darrow, and T. W. Morris, Noninvasive measurement of renal hemodynamic functions using gadolinium-enhanced magnetic resonance imaging, Invest. Radiol. 29, S123S126 1994.
19
M. H. Buonocore and R. W. Katzberg, Estimation of extraction fraction
EF and glomerular filtration rate GFR using MRI: Considerations derived from a new Gd-chelate biodistribution model simulation, IEEE
Trans. Med. Imaging 24, 651666 2005.
20
A. Thomas, M. Warm, M. Hoopmann, F. Diekmann, and T. Fischer, Tissue Doppler and strain imaging for evaluating tissue elasticity of breast
lesions, Acad. Radiol. 14, 522529 2007.
21
M. Sridhar, J. Liu, and M. F. Insana, Viscoelasticity imaging using ultrasound: Parameters and error analysis, Phys. Med. Biol. 52, 24252443
2007.
22
J. E. Drumm and R. ORahilly, The assessment of prenatal age from the
crown-rump length determined ultrasonically, Am. J. Anat. 148, 555
560 1977.
23
J. E. Drumm, J. Clinch, and G. MacKenzie, The ultrasonic measurement
of fetal crown-rump length as a method of assessing gestational age, Br.
J. Obstet. Gynaecol. 83, 417421 1976.
24
P. F. Judy, R. G. Swensson, and M. Szulc, Lesion detection and signalto-noise ratio in CT images, Med. Phys. 8, 1323 1981.
25
R. M. Nishikawa and M. J. Yaffe, Signal-to-noise properties of mammographic film-screen systems, Med. Phys. 12, 3239 1985.
26
W. A. Edelstein, P. A. Bottomley, and L. M. Pfeifer, A signal-to-noise
calibration procedure for NMR imaging systems, Med. Phys. 11, 180
185 1984.
27
D. P. Chakraborty and G. T. Barnes, Signal-to-noise ratio considerations
in radiographic imaging, Med. Phys. 10, 467469 1983.
28
B. Zhao, L. H. Schwartz, C. S. Moskowitz, M. S. Ginsberg, N. A. Rizvi,
and M. G. Kris, Lung cancer: Computerized quantification of tumor
responseinitial results, Radiology 241, 892898 2006.
29
S. Rohde, B. Turowski, J. Berkefeld, and A. F. Kovacs, CT-based evaluation of tumor volume after intra-arterial chemotherapy of locally advanced carcinoma of the oral cavity: Comparison with clinical remission
rates, Cardiovasc. Intervent. Radiol. 30, 8591 2007.
30
C. H. Holdsworth, R. D. Badawi, J. Manola, M. F. Kijewski, D. A. Israel,
G. D. Demetri, and A. D. Van den Abbeele, Use of CT and FDG-PET as
early prognostic indicators of response to imatinib mesylate Gleevec in
patients with gastrointenstinal stromal tumors GIST, AJR, Am. J.
Roentgenol. in press.
31
The IRAT network, http://www.aaci-cancer.org/irats/about network.asp,
accessed 2007.
32
J. Beuth and J. M. Schierholz, Review. Evidence-based complementary
oncology. Innovative approaches to optimize standard therapy strategies,
In Vivo 21, 423428 2007.
33
S. M. Erturk, S. Ondategui-Parra, H. Otero, and P. R. Ros, Evidencebased radiology, J. Am. Coll. Radiol. 3, 513519 2006.
34
D. L. Sackett, W. M. Rosenberg, J. A. Gray, R. B. Haynes, and W. S.
Richardson, Evidence based medicine: What it is and what it isnt.
1996, Clin. Orthop. Relat. Res. 455, 35 2007.
35
J. P. Ioannidis, Molecular evidence-based medicine, Eur. J. Clin. Invest.
37, 340349 2007.
36
G. W. Sledge, Jr., Evidence-based medicine in breast cancer, Clin.
Breast Cancer 7, 435 2007.
37
A. Fine, Medicare evaluates pay-for-performance, Manag. Care Q. 14,
2223 2006.
38
T. P. Meehan, Pay-for-performance: A new trend in health care, Conn.
Med. 71, 225226 2007.
39
R. Lu, P. Marziliano, and T. C. Hua, Liver tumor volume estimation by
semi-automatic segmentation method, Conf. Proc. IEEE Eng. Med. Biol.
Soc., 2005, Vol. 3, pp. 32963299.
40
P. J. Yim, A. V. Vora, D. Raghavan, R. Prasad, M. McAullife, P. OhmanStrickland, and J. L. Nosher, Volumetric analysis of liver metastases in
computed tomography with the fuzzy C-means algorithm, J. Comput.
Assist. Tomogr. 30, 212220 2006.
41
M. C. Ashe, K. M. Khan, S. A. Kontulainen, P. Guy, D. Liu, T. J. Beck,
and H. A. McKay, Accuracy of pQCT for evaluating the aged human
17
4179
radius: An ashing, histomorphometry and failure load investigation, Osteoporos. Int. 17, 12411251 2006.
42
B. C. Khoo, T. J. Beck, Q. H. Qiao, P. Parakh, L. Semanick, R. L. Prince,
K. P. Singer, and R. I. Price, In vivo short-term precision of hip structure
analysis variables in comparison with bone mineral density using paired
dual-energy x-ray absorptiometry scans from multi-center clinical trials,
Bone 37, 112121 2005.
43
L. J. Melton III, T. J. Beck, S. Amin, S. Khosla, S. J. Achenbach, A. L.
Oberg, and B. L. Riggs, Contributions of bone density and structure to
fracture risk assessment in men and women, Osteoporos. Int. 16, 460
467 2005.
44
P. D. Ross, Osteoporosis. Frequency, consequences, and risk factors,
Arch. Intern. Med. 156, 13991411 1996.
45
H. Kroger, M. Tuppurainen, R. Honkanen, E. Alhava, and S. Saarikoski,
Bone mineral density and risk factors for osteoporosisa populationbased study of 1600 perimenopausal women, Calcif. Tissue Int. 55, 17
1994.
46
M. E. Ooms, P. Lips, L. A. Van, and H. A. Valkenburg, Determinants of
bone mineral density and risk factors for osteoporosis in healthy elderly
women, J. Bone Miner. Res. 8, 669675 1993.
47
D. H. Miller et al., MRI outcomes in a placebo-controlled trial of natalizumab in relapsing MS, Neurology 68, 13901401 2007.
48
F. Fazekas, P. Soelberg-Sorensen, G. Comi, and M. Filippi, MRI to
monitor treatment efficacy in multiple sclerosis, J. Neuroimaging 17,
50S55S 2007.
49
M. A. Rocca and M. Filippi, Functional MRI in multiple sclerosis, J.
Neuroimaging 17, 36S41S 2007.
50
N. De Stefano and M. Filippi, MR spectroscopy in multiple sclerosis, J.
Neuroimaging 17, 31S35S 2007.
51
L. J. Thal, K. Kantarci, E. M. Reiman, W. E. Klunk, M. W. Weiner, H.
Zetterberg, D. Galasko, D. Pratico, S. Griffin, D. Schenk, and E. Siemers,
The role of biomarkers in clinical trials for Alzheimer disease, Alzheimer Dis. Assoc. Disord. 20, 615 2006.
52
M. Yoshita, E. Fletcher, D. Harvey, M. Ortega, O. Martinez, D. M. Mungas, B. R. Reed, and C. S. DeCarli, Extent and distribution of white
matter hyperintensities in normal aging, MCI, and AD, Neurology 67,
4179
21922198 2006.
D. E. Huddleston and S. A. Small, Technology insight: imaging amyloid
plaques in the living brain with positron emission tomography and MRI,
Nat. Clin. Pract. Neurol. 1, 96105 2005.
54
T. den Heijer, P. E. Sijens, N. D. Prins, A. Hofman, P. J. Koudstaal, M.
Oudkerk, and M. M. Breteler, MR spectroscopy of brain white matter in
the prediction of dementia, Neurology 66, 540544 2006.
55
E. M. Geraghty, J. M. Boone, J. P. McGahan, and K. Jain, Normal organ
volume assessment from abdominal CT, Abdom. Imaging 29, 482490
2004.
56
S. V. Raman, M. Shah, B. McCarthy, A. Garcia, and A. K. Ferketich,
Multi-detector row cardiac computed tomography accurately quantifies
right and left ventricular size and function compared with cardiac magnetic resonance, Am. Heart J. 151, 736744 2006.
57
P. Therasse, S. G. Arbuck, E. A. Eisenhauer, J. Wanders, R. S. Kaplan, L.
Rubinstein, J. Verweij, G. M. Van, A. T. van Oosterom, M. C. Christian,
and S. G. Gwyther, New guidelines to evaluate the response to treatment
in solid tumors. European Organization for Research and Treatment of
Cancer, National Cancer Institute of the United States, National Cancer
Institute of Canada, J. Natl. Cancer Inst. 92, 205216 2000.
58
J. M. Boone, Multidetector CT: Opportunities, challenges, and concerns
associated with scanners with 64 or more detector rows, Radiology 241,
334337 2006.
59
C. R. Meyer et al., Evaluation of lung MDCT nodule annotation across
radiologists and methods, Acad. Radiol. 13, 12541265 2006.
60
S. G. Armato III, G. McLennan, M. F. Nitt-Gray, C. R. Meyer, D.
Yankelevitz, D. R. Aberle, C. I. Henschke, E. A. Hoffman, E. A. Kazerooni, H. MacMahon, A. P. Reeves, B. Y. Croft, and L. P. Clarke, Lung
image database consortium: Developing a resource for the medical imaging research community, Radiology 232, 739748 2004.
61
M. H. Buonocore and R. W. Katzberg, Estimation of extraction fraction
EF and glomerular filtration rate GFR using MRI: Considerations derived from a new Gd-chelate biodistribution model simulation, IEEE
Trans. Med. Imaging 24, 651666 2005.
62
Imaging as a biomarker: Standards for change measurements in therapy,
2007.
53
Received 1 August 2007; revised 9 October 2007; accepted for publication 13 October 2007;
published 28 November 2007
A favorite clich, often attributed to Yogi Berra, is Prediction is difficult, especially about the
future! In this brief review, we shall recall some historical difficulties in predicting the future of
diagnostic medical imagingin particular, the modern technologies of CT and MRIand examine
the analogous situation in the emerging technology of diagnostic microarrays for the multiplebiomarker problem. All of these technologies began their lives as ill-conditioned problems, i.e.,
there was insufficient data to solve the central problem at hand. DOI: 10.1118/1.2805252
I. OVERVIEW
In the first half of this article we will look at how well investigators in the past predicted the future of technology in
diagnostic medical imaging, and their vulnerability to a wellknown dictum about forecasting.1 In the second half we will
consider an analogous situation for the new kind of medical
image represented by genomic and proteomic microarrays.
These latter emerging technologies allow for simultaneous
measurementfrom a single tissue or blood sampleof the
levels of expression of many thousands of different gene or
protein fragments using a novel chip design that merges molecular biology and optoelectronics. We will find some natural parallels between diagnostic imaging and microarrays. At
the end we will identify a central theme from the assessment
of conventional medical imaging that can offer insight into
the evolution of contemporary microarray technologies.
The topics covered here were selected from the research
experience of the author and his professional colleagues because they exemplify the difficulty of predicting the course
of the history of these technologies while it was being lived
in real time. No attempt has been made to provide a broad
history of either the very large field of medical imaging or
that of multiple-biomarker microarray technologies.
First, let us remark that a medical image is a random array
or vector: Random because there are multiple sources of
variability or randomness, including detector noise and interpatient and intrapatient variability, and array because an
image is obviously a multidimensional matrix. Multivariate
statistics is therefore an essential tool for understanding the
amount of information that can be captured in a medical
image. The new technology of multiple-biomarker microarrays may be thought of as a special category of medical
imaging. In conventionally processed medical image arrays,
the underlying relational structures are usually obvious because natural morphology and function are typically displayed directly. In the case of microarrays, computationally
intensive statistical techniques are necessary to discover any
relational structure in the data and within and across samples
from nondiseased and diseased patients. An obvious limitation of microarrays is that they do not generally localize their
findings to well-specified sites in the body. So medical im4944
Let us consider some critical moments during the emergence of CT and magnetic resonance imaging MRI during
the 1970s. Recall that these technologies were both enabled
by giant steps in digital technologies, especially the minicomputer, the computer in a breadbox that for the first time
allowed complex mathematical image reconstruction algorithms to be executed on large format images in a manageable time and space and therefore with manageable expense.
While the mathematical algorithm community has naturally emphasized the open-ended potential for their own specialized contributions to imaging, physicists have tried to
understand not only the potential, but also the limitations,
imposed on these technologies by the laws of physics and
statisticsin particular, the quantity and quality of the
samples of input data required for clinically useful images.
0094-2405/2007/3412/4944/8/$23.00
4944
4945
4945
differences.47 It took some years before a widespread understanding emerged that CT was in principle quantum limited,
and thus highly efficient as defined above, for low-contrast
tasks. Investigators who wished to push the envelope in this
field then turned their attention to the question of whether
reductions in the number of views could be efficiently taken
for certain high-contrast imaging applications or by the incorporation of prior information and constraints.8
III.C. The early predictions for MRI
FIG. 1. Example of an early CT image of the brain showing a large spacefilling lesion. The conditioning of this inverse problem can be improved to
some extent by simply demanding lower resolution in the image, achievable
most simply by blurring ones vision. Courtesy of Siemens Medical
Solutions.
Both CT and MRI are, in principle, mathematically illconditioned because they attempt to solve an inverse problem of reconstructing a continuous object i.e., the body
from a finite number of discrete and noisy data samples.2 The
original and most common way of dealing with this illconditioning is by relaxing the resolution requirements on
the final result until a useful image, with disguised or hardly
visible artifacts, is obtained see, for example, Fig. 1. For
our present purposes, we define an artifact as a structure in
the image not present in the original object and not due
solely to random photon or detector noise.
One of the principal benchmarks used for assessing these
imaging technologies is the ability to discriminate a region of
a given size and contrast from its surround, versus the radiation dose or exposure and/or imaging time required for this
task. We may refer to this benchmark of image quality
versus dose or exposure and/or imaging time as imaging efficiency. How did the early investigators view the prospects
for imaging efficiency in CT and MRI?
III.B. The early predictions for CT
Some of the early investigators of MRI or nuclear magnetic resonance NMR imaging as it was then called were
pessimistic about achieving clinically useful levels of resolution in realistic imaging times. Signal-to-noise SNR calculations based on elementary theories of signal detection
and statistical decision making lead to the conclusion that
imaging time in three dimensions depends on the inverse
sixth power of the resolution required or fourth power in
two dimensions, among other variations of the scenario9. I
attended an MRI brainstorming meeting at NIH in the late
1970s. David Hoult, a pioneer in the field who was recruited
by NIH to develop an early clinical scanner for them, emphasized his concern that attempts to reduce the limiting
resolution of MRI would face a formidable obstacle from the
constraint of the power law just cited. In a few words, if
image resolution was to be improved by a factor of 2, the
imaging time might have to be increased by a factor ranging
from 16 to 64, and this would not be clinically practical.
On May 45, 1995 I attended a meeting at the National
Library of Medicine in celebration of the centenary of the
discovery of x rays by Roentgen Radiology Centennial
Commemorative Conference. Pioneers as well as contemporary experts across many fields of medical imaging presented
their experience of the progress in their fields. One of the
presenters was a major player in the development of both CT
and MRI at General Electric Corporate Research and Development Laboratories, Lewis S. Lonnie Edelheit, who had
succeeded the beloved mentor of many of us of the previous
generation, Rowland W. Red Redington. In Edelheits presentation to this commemorative conference he showed a
slide containing a copy of a letter he had written to GE
management early in the development of MRI. The letter
told a story similar to that of Hoult, namely, the outlook for
improved resolution in MRI was not great owing to the
strong dependence of imaging time on required resolution;
he was not able to encourage GE management to increase
their support for development of MRI at that point.
A brief historical summary of the evolution of that view
was presented by Redington in Ref. 10. Redington emphasized that, among other points, the previous discussions on
resolution neglected to distinguish between high- and lowcontrast regimes and ignored the great soft-tissue contrast
available in MRI. His short paper also includes a discussion
of the difference between signal, noise, and resolution characteristics in MRI versus in CT that guided some of our own
work.9 He outlined in vivo NMR spectroscopy as a potential
answer to what comes next after NMR imaging?. And he
4946
4946
demand forsteady miniaturization and increase in the number of CT detectors, both circumferentially around the body
and axially along the body. In the early days of CT, many
investigators thought that such an increase would automatically lead to an increase in required x-ray dose. However, the
large-area low-contrast performance may be maintained independently of the number of detectorsit is not the number
of photons per detector that directly determines the dose/
image-quality relationship in the low-contrast regime, it is
rather the spatial density of detected x-ray counts9as long
as the number of detected counts exceeds the dark noise and
readout noise of the detector.
A major unsurprising and long-awaited development has
been the emergence of technologies foreseen more than three
decades ago but only made practical by advances in digital
detectors, e.g., dual-energy x-ray imaging and limited-angle
tomography or tomosynthesis. One popular prediction gradually being realized is that many if not most 2D or planar
imaging technologies will be supplanted by 3D volumetric
imaging technologies in the foreseeable future. The radiation
dose to the body may increase appreciably for some of these
latter developments.
Finally, an unconventional re-examination of the geometry of CT scanning has led to the investigation of a configuration referred to as inverse-geometry volumetric CT.15 The
name derives from the fact that a large-area, scanned x-ray
source is used with a detector array that is smaller than the
source in the transverse direction but comparable in the
axial direction. The authors claim that noise and resolution
with their micro-CT system are comparable to an existing
volumetric micro-CT system, while avoiding the cone-beam
artifacts that occur in the latter. Additional projected benefits
include reduced scatter and detector cost.
To summarize, the first generations of investigators in
both CT and MRI did not have a clear view of the requirements for the number of image samples, e.g., photons and
radiation exposure or dose, views, and imaging time, needed
for clinically useful images for particular imaging tasks.
Much work advanced on an ad hoc basis, in particular by
means of a beauty contest between competing modalities
or algorithmscontinuing in this mode up to the present. A
foundational approach to many of the issues sketched above
required several decades to formulate and has been published
in the present decade by Barrett and Myers.2 This approach
should appeal naturally to the physicists in our audience. It
depends in part on physical measurement and analysis of the
so-called singular vectors, or natural imaging building
blocks corresponding to a given physical configuration of
image detection system; this analysis leads to a separation
between the concepts of the measurement space and the
null space. The measurement space is that portion of the
object that is accessible to measurement by the imaging system; the null space is that portion of the object that is inaccessible to measurement by the imaging system. A trivial
example of a null space would be the zeroes of the system
transfer function. Thus, objects that differ only in their nullspace component appear the same when imaged. Many artifacts may be thought of as ghosts from the null space,8 i.e.,
4947
The biomedical landmark of our time has been the sequencing of the genomes of many organisms, in particular
the sequencing of the human genome. As more genes and
proteins are identified, and those involved in disease initiation and progression become better understood, an increasing
number of candidate diagnostic biomarkers as well as novel
therapeutic agents are expected to emerge.19 The target of
the diagnostic and therapeutic agents may be the general
population, or it might be some specially targeted subpopulations. The targeting of therapeutics based on the biological
make-up of subclasses of patients is known as pharmacogenomics and is popularly referred to as personalized medicine. The promise of the field is great and open-ended,
yetas was the case with the breakthrough technologies in
medical imagingthe scale of the promise and efficiency
requirements for developing clinically useful diagnostic tools
is not yet well understood.
Investigators emphasize that it is essential to distinguish
between genetics and genomics. In genetics, one tests for the
presence or absence or usually the mutation status of one or
typically only a few specified genes, e.g., the gene associated
with cystic fibrosis or the BRCA1 and BRCA2 genes assoMedical Physics, Vol. 34, No. 12, December 2007
4947
ciated with a class of breast cancers. In genomics, proteomics, and related technologies, however, investigators are testing not for the presence or absence, but rather for the level of
expression, of a large number of genes and/or proteins, and
patterns of expression across many genes or protein fragments. The physical measurements in the proteomic approach are obtained from mass spectrometry of specially prepared tissue specimens. The physical measurements in the
genomic approach will be sketched here for interested newcomers. Both technologies are complicated by potential
variations in expression level as a function of time of day or
metabolic or other natural cycle including, of course, the
patients age.
The technology used in genomics to measure the level of
expression of a large number of specified genes simultaneously is referred to as a microarray assay; an individual
microarray is often referred to as a DNA chip and is about
the size of a thumbprint. There are several different contemporary microarray technologies.20 We provide here an introductory summary of the generic approach. A microarray contains a location and spot for each of the specified reference
genes. The spots can be prepared by synthesizing previously
designated sections of known genetic sequence and binding a
quantity of this material to a substrate. The spot, together
with some associated controls, is referred to as a probe. In
contemporary work, the number of probes on a chip is in the
order of tens of thousands. A prepared sample of genetic
material from a patients blood or other tissue of interest, the
target, is labeled with a fluorescent dye, and then washed
over the chip. The goal of the measurement is to quantify the
degree of hybridization between the material in the target
and that in the probe. Hybridization refers to the complementary binding of the bases in the genetic code A to T, G to C,
etc.. The degree of hybridization at each spot is assumed to
be proportional to the number of copies of the specified RNA
section in the patients tissue samplea measure of the level
of expression of the specified gene. After hybridization, the
spots on the microarray are read out using laser light to excite the fluorescence, optics and photodetection to measure
the fluorescence, and finally digitization. Microarrays are
thus a form of molecular imaging of tissue samples extracted
from patients.
As in medical imaging but greatly more so, the genomics and proteomics problems are not generally well conditioned; great variability in data consistent with a wide range
of potential but not real solutions to any particular diagnostic problem can arise at each of several stages of analyzing
the problem. The very first stage involves quality control of
the laboratory conditions and measurement apparatus. There
are a multitude of issues here, well beyond our present scope.
The next principal stagetrying to find those genes that are
biologically significant for a particular task, among many
thousands of candidatesis fraught with the general problems of all tasks of data mining, namely, the fact that many
candidates are expected to stand out in a noisy sample purely
by random chance, so-called false discovery.20 Newcomers
may be surprised to learn that a typical starting point for the
overall procedure is measurement of the level of expression
4948
A crucial final stage involves the task of training a classifier how to weight the levels of expression of a modest number of gene or protein fragments that survive the initial data
mining, using a statistical learning machine, e.g., a classical or neural-network classifier, to maximize success in classifying the condition of interest, e.g., cancer versus noncancer, or an aggressive subclass of cancer versus an indolent
one, or a responder versus a nonresponder to a specified
therapy, among others. A natural bottleneck occurs here since
at present the accessible and affordable world provides only
a finite sample of patients with known truth status who can
be used to train the classifier, and next to test it with previously unseen cases. This limitation of samples can lead to
great uncertainty in the classifier itself, as well as attempts to
estimate its performance over the target population using any
modest number of available patients.
In general, the uncertainty in the performance estimate
decreases with increases in either or both of the number of
training samples and the number of testing samples. The issues here are conceptually identical to those one encounters
when studying task-dependent trained model observers when
assessing medical imaging systems.21 As in this latter problem, the uncertainty in performance estimation also decreases with increasing intrinsic separability of the populations. This is analogous to the intrinsic contrast of a
particular diagnostic task in medical imaging. A further issue
for the problem of statistical learning machines is that the
number of training samples required for a given confidence
level also increases with the number of biomarkers features
in the classifier and/or the complexity of the classifier.22
Because of the great opportunity for false discovery of
genes at the early stage of development of a given technology and the limitations of available samples from patients
with known truth status, many early promises have not held
up under later scrutiny, while the jury is still out on many
other high-profile studies.
The work of Golub and colleagues on classification of
human acute leukemias23 has attracted a great deal of attention for several reasons, including the fact that it was one of
Medical Physics, Vol. 34, No. 12, December 2007
4948
4949
4949
well as a new random sample of testers? Of course, confidence intervals for the latter approach will in general be
wider than those for the former approach.
The point of view of myself and collaborators is that the
second, or more general, uncertainty should be specified.28,29
Classifier designers sometimes argue that their training is
frozen. However, it is almost always the intent of these
investigators to continue to build their database, improve
their statistical learning, and update their algorithms. This is
motivation for the more general approach of Refs. 28 and 29.
The former may be useful for analyzing a pilot study to
guide the sizing of a larger subsequent pivotal study; the
latter can apply equally to the setting of either a pilot or a
pivotal study. An analysis of the uncertainties in a pilot study
may be the only way to design a more definitive pivotal
study.
Further motivation for our more general position can be
drawn from a recent exchange in the multiple-biomarker literature. Dave and colleagues31 trained a model for
prediction-of-survival on 95 biopsy specimens from patients
4950
4950
4951
101 2000.
K. K. Vigen, D. C. Peters, T. M. Grist, W. F. Block, and C. A. Mistretta,
Undersampled projection-reconstruction imaging for time-resolved
contrast-enhanced imaging, Magn. Reson. Med. 43, 170176 2000.
13
C. A. Mistretta, O. Wieben, J. Velikina, W. Block, J. Perry, Y. Wu, K.
Johnson, and Y. Wu, Highly constrained backprojection for timeresolved MRI, Magn. Reson. Med. 55, 3040 2006.
14
C. A. Mistretta, Innovative physics boosts MRI speed, cuts CT dose,
Invited article for Diagnostic Imaging magazine August 2007, pp. 33
42, concluded on p. 54.
15
T. G. Schmidt, J. Star-Lack, N. R. Bennet, S. R. Mazin, E. G. Solomon,
R. Fahrig, and N. J. Pelc, A prototype table-top inverse-geometry volumetric CT system, Med. Phys. 33, 18671878 2006.
16
R. F. Wagner, Lessons from my Dinners with the Giants of Modern
Image Science, Radiat. Prot. Dosimetry 114, 410 2005.
17
K. J. Myers, R. F. Wagner, K. M. Hanson, H. H. Barrett, and J. P. Rolland,
Human and quasi-Bayesian observers of images limited by quantum
noise, object variability, and artifacts, Proc. SPIE 2166, 180190 1994.
18
Details of many applications of the approach of Ref. 17 may be found in
manuscript or abstract form on the website of K. M. Hanson at Los
Alamos, http://public.lanl.gov/kmh/publications
19
G. S. Burrill, Biotech 200620th Anniversary Report on the Industry.
Life Sciences: A Changing Prescription Burrill and Company, San Francisco, CA, 2006.
20
G. J. McLachlan, K.-A. Do, and C. Ambroise, Analyzing Microarray
Gene Expression Data Wiley, Hoboken, NJ, 2004.
21
B. D. Gallas, Estimating the variance of the ideal linear observers
signal-detection performance from a finite number of trainers and testers,
presented at Medical Image Perception Conference X, Duke University,
Durham, North Carolina 2003. Proceedings of the conference on disc
available from Medical Imaging Perception Society MIPS, or file available from [email protected]
22
V. N. Vapnik, The Nature of Statistical Learning Theory Springer-Verlag,
New York, 1995.
23
T. R. Golub et al., Molecular classification of cancer: Class discovery
and class prediction by gene expression monitoring, Science 286, 531
537 1999.
24
T. M. Cover, Geometrical and statistical properties of systems of linear
inequalities with applications in pattern recognition, IEEE Trans. Electron. Comput. EC-14, 326334 1965.
25
E. F. Petricoin et al., Use of proteomic patterns in serum to identify
ovarian cancer, Lancet 359, 572577 2002.
12
26
4951
D. E. Ransohoff, Bias as a threat to the validity of cancer molecularmarker research, Nat. Rev. Cancer 5, 142149 2005.
27
W. A. Yousef, R. F. Wagner, and M. H. Loew, Comparison of nonparametric methods for assessing classifier performance in terms of ROC
parameters, in Applied Imagery Pattern Recognition Workshop, 2004:
Proceedings of the 33rd AIPR Conference IEEE Computer Society, Los
Alamitos, CA, 2004, pp. 190195.
28
W. A. Yousef, R. F. Wagner, and M. H. Loew, Estimating the uncertainty
in the estimated mean area under the ROC curve of a classifier, Pattern
Recognit. Lett. 26, 26002610 2005.
29
W. A. Yousef, R. F. Wagner, and M. H. Loew, Assessing classifiers from
two independent data sets using ROC analysis: A nonparametric approach, IEEE Trans. Pattern Anal. Mach. Intell. 28, 18091817 2006
TPAMI.
30
W. A. Yousef, R. F. Wagner, and M. H. Loew, The partial area under the
ROC curve: Its properties and nonparametric estimation for assessing
classifier performance, submitted to IEEE Trans. Pattern Anal. Mach.
Intell., 2007.
31
S. S. Dave et al., Prediction of survival in follicular lymphoma based on
molecular features of tumor-infiltrating immune cells, N. Engl. J. Med.
351, 21592169 2004.
32
R. Tibshirani, Immune signatures in follicular lymphoma, N. Engl. J.
Med. 352, 14961497 2005.
33
H. P. Chan, B. Sahiner, R. F. Wagner, and N. Petrick, Classifier design
for computer-aided diagnosis: Effects of finite sample size on the mean
performance of classical and neural network classifiers, Med. Phys. 26,
26542668 1999.
34
B. Sahiner, H. P. Chan, N. Petrick, R. F. Wagner, and L. Hadjiiski, Feature selection and classifier performance in computer-aided diagnosis:
The effect of finite sample size, Med. Phys. 27, 15091522 2000.
35
R. F. Wagner, C. E. Metz, and G. Campbell, Assessment of medical
imaging systems and computer aids: A Tutorial Review, Acad. Radiol.
14, 723748 2007.
36
S. V. Beiden, M. A. Maloof, and R. F. Wagner, A general model for
finite-sample effects in training and testing of competing classifiers,
IEEE Trans. Pattern Anal. Mach. Intell. 25, 15611569 2003.
37
R. M. Simon, E. L. Korn, L. M. McShane, M. D. Radmacher, G. W.
Wright, and Y. Zhao, Design and Analysis of DNA Microarray Investigations Springer-Verlag, New York, 2004.
38
D. Gur, R. F. Wagner, and H.-P. Chan, On the repeated use of databases
for testing incremental improvement of computer-aided detection
schemes, Acad. Radiol. 11, 103105 2004.
Received 26 August 2007; revised 2 October 2007; accepted for publication 1 November 2007;
published 19 December 2007
A brief history of the underlying principles of the conventional fractionation in radiation therapy is
discussed, followed by the formulation of the hypothesis for hypofractionated stereotactic body
radiation therapy SBRT. Subsequently, consequences of the hypothesis for SBRT dose shaping
and dose delivery techniques are sketched. A brief review of the advantages of SBRT therapy in
light of the existing experience is then provided. Finally, the need for new technological developments is advocated to make SBRT therapies more practical, safer, and clinically more effective. It
is finally concluded that hypofractionated SBRT treatment will develop into a new paradigm that
will shape the future of radiation therapy by providing the means to suppress the growth of most
carcinogen-induced carcinomas and by supporting the cure of the disease. 2008 American Association of Physicists in Medicine. DOI: 10.1118/1.2816228
Key words: ablative radiation therapy, hypofractionation, SBRT
I. INTRODUCTION
I.A. Historical foundation of conventional fractionation
schemes
Traditional radiation therapy delivery requires multiple fractions of 1.5 to 3 Gy administered daily over a period of 37
weeks. This regimen has been established based on early
experience of treatment with radiation and has been justified
by widely accepted models of radiobiological effects of x
rays on human tissue.13 However, the strategy of traditional
fractionation in external beam radiation therapy has not been
the only one practiced historically. After the discovery of
ionizing radiation, hypofractionated, or even single large
dose fraction treatments, were practiced. It has been observed that large doses per fraction were tumorcidal, especially for epithelial tumors. Furthermore, early clinical experience provided valuable lessons regarding the balance
between tumor control and tissue toxicities. Early evaluations of radiation therapy showed that the delivery of a large
dose per fraction treatments was leading to unacceptably
high toxicities. Therefore, quite early in the development of
radiation therapy these schedules were abandoned because of
complications such as fibrosis, stenosis, and vascular injury.
Data collected at initial stages of the development of radiation therapy were irrefutable. The understanding of the
factors contributing to complications was incomplete. The
ability to manage the parameters underlying the unfavorable
results for large dose fraction therapy was severely limited in
the early days of radiation therapy by immature technologies
of dose delivery. However, these early experiences and their
impact on the acceptance of particular fractionation schemes
have been largely forgotten by later practitioners of radiation
therapy. The paradigm of delivering a dose in small portions
has been taken for granted and disconnected from the reflection that early treatments suffered multiple dosimetric limitations. These limitations included, among others, the inability to decrease exposure of normal tissues due to low beam
112
0094-2405/2008/351/112/7/$23.00
112
113
assured under the provision of multiple, conventional fractionation. Finally, since oxygenated tissue was considered to
be more radiosensitive, the better reoxygenation of tumors
during multifractionated dose delivery was thought to be advantageous for the therapy.
The overall mechanism of positive differential effects in
eradication of tumor cells vs normal tissues in radiation
therapy based on multifractionation regimes has been called
the therapeutic window. However, the therapeutic window
benefits have not always been well documented and sometimes actually may not have worked very well for the therapeutic advantage. In particular, all the above-described
mechanisms could, under specific conditions, act to the disadvantage of radiation therapy. For example, tumors have a
capacity to repair and tumors may repopulate too. In turn,
normal tissues can also become more sensitive throughout
the course of radiotherapy. Thus, under certain circumstances
and for certain types of cancers and treatment sites, fractionation may have actually been detrimental rather than advantageous for the success of radiation therapy treatment.
I.C. The conventional fractionation vs Gamma Knife
experience
113
114
114
115
The precision of the setup and delivery required for hypofractionated therapies makes technologies of IMRT and
IGRT though not indispensable as explained above well
suited for efficiently carrying out SBRT treatments. In turn,
the SBRT therapy may be specifically well suited for showing the value of these new technologies to cancer therapy.
The clinical gains resulting from IMRT and IGRT may be
difficult to prove for standard fractionations. For traditional
radiation therapy, advances in local control are not expected
to be dramatic when IMRT and IGRT are implemented, and
improved survival data would be notoriously difficult to
demonstrate. However, in the case of SBRT treatment, the
combination of radical dose fractionation and the ability to
contain the toxicity, inured by superior accuracy of technologically advanced delivery, will allow more ready measurement of the improvements in local control and in survival
rates. Therefore, the overall results of SBRT delivery, when
supported by IMRT and IGRT technology, should establish
these technologies clinical credibility. However, as will be
Medical Physics, Vol. 35, No. 1, January 2008
115
III. DISCUSSION
III.A. SBRTvision for future
116
116
117
117
for stage I non-small cell lung cancer, Lung Cancer 48, 107114 2005.
P. Fritz et al., Stereotactic, single-dose irradiation of stage I non-small
cell lung cancer and lung metastases, Radiat. Oncol. 1, 30 2006.
24
H. Hof et al., Stereotactic single-dose radiotherapy of stage I non-smallcell lung cancer NSCLC, Int. J. Radiat. Oncol., Biol., Phys. 56, 335
341 2003.
25
U. Haedinger, W. Thiele, and J. Wulf, Extracranial stereotactic radiotherapy: Evaluation of PTV coverage and dose conformity, Med. Phys.
12, 221229 2002.
26
U. Haedinger, J. Wulf, U. Oppitz, B. Olshausen, W. Thiele, and M.
Flentje, Stereotactic treatment of extracranial targets: Evaluation of planning target volume PTV to compensate for target mobility due to target
motion and inaccuracy of patient positioning *1 , Int. J. Radiat. Oncol.,
Biol., Phys. 48, 346347 2000.
27
U. Haedinger, J. Wulf, U. Oppitz, W. Thiele, and M. Flentje, Extracranial stereotactic radiotherapy: Evaluation of the PTV to compensate target
mobility and set-up inaccuracy over the complete target volume measured
by DVH from repeated CT-simulation, Int. J. Radiat. Oncol., Biol., Phys.
51, 4546 2001.
28
M. Nevinny-Stickel, R. A. Sweeney, R. J. Bale, A. Posch, T. Auberger,
and P. Lukas, Reproducibility of patient positioning for fractionated extracranial stereotactic radiotherapy using a double-vacuum technique,
Strahlenther. Onkol. 180, 117122 2004.
29
T. Xia et al., Promising clinical outcome of stereotactic body radiation
therapy for patients with inoperable Stage I/II non-small-cell lung cancer, Int. J. Radiat. Oncol., Biol., Phys. 66, 117125 2006.
30
Y. Nagata et al., Clinical outcomes of a phase I/II study of 48 Gy of
stereotactic body radiotherapy in 4 fractions for primary lung cancer using a stereotactic body frame, Int. J. Radiat. Oncol., Biol., Phys. 63,
14271431 2005.
31
R. Hara et al., Clinical outcomes of single-fraction stereotactic radiation
therapy of lung tumors, Cancer 106, 13471352 2006.
32
H. Onishi et al., Stereotactic hypofractionated high-dose irradiation for
stage I nonsmall cell lung carcinoma: Clinical outcomes in 245 subjects
in a Japanese multi-institutional study, Cancer 101, 16231631 2004.
33
R. Timmerman et al., Extracranial stereotactic radioablation: Results of
a phase I study in medically inoperable stage I non-small cell lung cancer, Chest 124, 19461955 2003.
34
L. E. Gaspar and T. E. Schefter, Analysis of extracranial stereotactic
radiosurgery via equivalent uniform dose and tumor control probability:
Practical ramifications and translational implications, Int. J. Radiat. Oncol., Biol., Phys. 54, 225 2002.
35
B. D. Kavanagh, R. D. Timmerman, S. H. Benedict, Q. Wu, T. E.
Schefter, K. Stuhr, S. McCourt, F. Newman, R. M. Cardinale, and L. E
Gaspar, How should we describe the radioblologic effect of extracranial
stereotactic radlosurgery: Equivalent uniform dose or tumor control probability? Med. Phys. 30, 321324 2003.
36
C. R. Kelsey, T. Schefter, R. Nash, P. Russ, A. E. Baron, C. Zeng, and L.
F. Gaspar, Retrospective clinicopathologic correlation of gross tumor
size of hepatocellular carcinoma: Implications for extracranial stereotactic
radiosurgery, Int. J. Radiat. Oncol., Biol., Phys. 57, S283 2003.
37
L. Papiez, R. Timmerman, C. DesRosiers, and M. Randall, Extracranial
stereotactic radioablation: Physical principles, Acta Oncol. 42, 882894
2003.
38
J. E. Reiff, M. Werner-Wasik, P. R. Anne, R. K. Valicenti, and J. M.
Galvin, Interfraction variations of the isocenter relative to coordinates of
a stereotactic body frame for the treatment of extracranial lesions, Int. J.
Radiat. Oncol., Biol., Phys. 48, 345 2000.
39
J. E. Reiff, M. Werner-Wasik, B. E. Garofola, R. K. Valicenti, P. R. Anne,
A. P. Dicker, W. J. Curran, Jr., and J. M. Galvin, The efficacy of a
stereotactic body frame in an adaptive radiotherapy program, Int. J. Radiat. Oncol., Biol., Phys. 51, 172173 2001.
40
R. D. Timmerman, L. Papiez, R. McGarry, L. Likes, C. DesRosiers, S.
Frost, and M. Williams, Extracranial stereotactic radioablation: Results
of a phase I study in stage I non-small cell lung cancer, Int. J. Radiat.
Oncol., Biol., Phys. 57, S280S281 2003.
41
R. Timmerman, L. Papiez, R. McGarry, L. Likes, C. DesRosiers, S. Frost,
and M. Williams, Extracranial stereotactic radioablation: Results of a
phase I study in medically inoperable stage I non-small cell lung cancer,
Chest 124, 19461955 2003.
42
R. Timmerman, L. Papiez, and M. Suntharalingam, Extracranial stereotactic radiation delivery: Expansion of technology beyond the brain,
Technol. Cancer Res. Treat. 2, 153160 2003.
23
118
43
118
George X. Ding
Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
Anders Ahnesj
Department of Oncology, Radiology and Clinical Immunology, Section of Oncology,
Uppsala University, S-751 85 Uppsala & Nucletron AB, S-751 47 Uppsala, Sweden
Received 31 July 2007; revised 24 September 2007; accepted for publication 18 October 2007;
published 20 December 2007
Advances in radiation treatment with beamlet-based intensity modulation, image-guided radiation
therapy, and stereotactic radiosurgery including specialized equipments like CyberKnife, Gamma
Knife, tomotherapy, and high-resolution multileaf collimating systems have resulted in the use of
reduced treatment fields to a subcentimeter scale. Compared to the traditional radiotherapy with
fields 4 4 cm2, this can result in significant uncertainty in the accuracy of clinical dosimetry.
The dosimetry of small fields is challenging due to nonequilibrium conditions created as a consequence of the secondary electron track lengths and the source size projected through the collimating
system that are comparable to the treatment field size. It is further complicated by the prolonged
electron tracks in the presence of low-density inhomogeneities. Also, radiation detectors introduced
into such fields usually perturb the level of disequilibrium. Hence, the dosimetric accuracy previously achieved for standard radiotherapy applications is at risk for both absolute and relative dose
determination. This article summarizes the present knowledge and gives an insight into the future
procedures to handle the nonequilibrium radiation dosimetry problems. It is anticipated that new
miniature detectors with controlled perturbations and corrections will be available to meet the
demand for accurate measurements. It is also expected that the Monte Carlo techniques will increasingly be used in assessing the accuracy, verification, and calculation of dose, and will aid
perturbation calculations of detectors used in small and highly conformal radiation beams. 2008
American Association of Physicists in Medicine. DOI: 10.1118/1.2815356
Key words: small field dosimetry, nonequilibrium conditions, Monte Carlo
I. INTRODUCTION
With image guidance and improved treatment delivery techniques at the core of modern radiation therapy, the treatment
fields that traditionally spanned from 4 4 cm2 up to 40
40 cm2 are now being reduced down to a subcentimeter
range in advanced and specialized radiation treatments such
as beamlet-based intensity modulated radiation therapy
IMRT, image-guided radiation therapy IGRT, tomotherapy, stereotactic radiosurgery SRS with high-resolution
multileaf collimator, Gamma Knife, and CyberKnife. In particular, the SRS, Gamma Knife, and CyberKnife rely on very
small field sizes on the order of a few millimeters to treat
tumors and spare normal structures. IMRT requires the addition of small fields with nonequilibrium conditions to treat
target volumes using optimization routines. Several dosimetric challenges due to this trend are lack of charged particle
equilibrium CPE,1,2 partial blocking of the beam source
giving rise to pronounced and overlapping penumbra,36 the
availability of small detectors for sizes comparable to the
field dimensions,7,8 and variations of the electron spectrum
inducing changes in stopping power ratios.9,10
Electronic equilibrium is a phenomenon associated with
the range of secondary particles and hence dependent on the
206
beam energy, the composition of the medium, and particularly the density of the medium. With a large variety of radiation detectors marketed by various manufacturers covering all sizes from mini to micro, types ionization chamber,
semiconductor, chemical, film, etc., and shapes thimble,
spherical, plane parallel, the choice of a suitable detector for
small field dosimetry could be a challenging and rather confusing task without proper guidelines. It is not too uncommon in clinical practice to compare measurements with various detectors and choose the detector that yields the highest
output for a given field size, or to select a measured value
that is common to several detectors, without proper consideration of the possible perturbations and corrections for each
of the detectors. Such approaches do not provide the scientific basis needed to achieve the confidence for dosimetric
accuracy commonly set for clinical practices. To deal with
these difficulties, a growing number of authors1017 have reported the comparison of measured data with Monte Carlo
simulation. However, at the present time Monte Carlo simulations cannot be assumed to invariably provide a gold standard without appropriate experimental validation. These developments challenge the conventional way of performing
dosimetric measurement and treatment planning. The pur-
0094-2405/2008/351/206/10/$23.00
206
207
207
pose of this paper is to illustrate the problem, provide possible solutions, and predict future trends in small field radiation dosimetry.
II. PROBLEMS
II.A. What is small?
For photon beams, measured data are used in dose calculation to provide the absolute dose normalization at a reference field, and through a beam modeling procedure indirectly drive dose calculations based on relative data such as
total scatter factor Scp, tissue maximum ratio TMR, percent depth dose PDD, and off-axis ratio OAR. The refer-
208
208
FIG. 3. The graph shows the primary dose profile in water across a collimating edge for different beam energies, specified by their quality index
TPR20/10, ranging from 0.68 to 0.80 at an interval of 0.2. Ideal spot
source fluence profiles are assumed with only the electron transport that
contributes to the penumbra width.
DtE,r
Dtr
=
Dtref
Dtref
FIG. 2. The effects of source size and beam shaping geometry on the output
of a small field, a 6 6 mm2 and b 24 24 mm2. Both field sizes are
defined by the micro multi-leave collimator mMLC and the variation are
caused by different settings of auxilary jaws. Reproduced with permission
from Ding et al., Ref. 21.
ence dose calibration is performed according to the dosimetry protocols24,25,27 with a well-defined beam geometry,
where beam quality and dosimetric parameters are known to
a high degree of accuracy. Dose measurements with ionization chambers rely on the assumptions of cavity theory.
When the size of the cavity is smaller than the range of
charged particles originated in the medium, the cavity is
treated as nonperturbing. In such a situation, the dose to the
medium is related to the dose to the air in the cavity by the
stopping power ratios of medium to air. However, with decreasing field size, neither the CPE nor the conditions for the
cavity theory can be fulfilled due to the lateral range of the
electrons. For a small field when the CPE does not exist, the
presence of a detector can change the local level of the CPE,
adding more perturbations to complicate the problem.
Under electronic equilibrium, cavity theory describes a
method to calculate the dose D in a medium based on
measured charge in the cavity,
Q
Dt =
m
W
e
,
a
W
e
ref
a ref
209
209
from the RPC values have been observed, indicating the seriousness of the dosimetric problem. The accurate dosimetry
for small field remains a problem until an appropriate and
consistent procedure is available and widely accepted by a
majority of users to calibrate nonstandard beams.
II.G. Low-density inhomogeneity
In low-density medium like the lung, small fields are subject to significant perturbations that are energy- and density
dependent. Treatment planning algorithms that use simple
one-dimensional density scaling fail to provide accurate dose
distributions as noted in several publications.4551 Advanced
treatment planning algorithms in general provide more accurate dose calculations in treatment planning.34,5254 However,
in clinical trials, a consistent approach in radiation dosimetry
is necessary. In order to make the outcome comparison
meaningful, new treatment protocols have yet to take advantage of these more accurate treatment planning algorithms,
which are now available in many commercial 3D treatment
planning systems.
III. PRESENT KNOWLEDGE
III.A. Experiment
210
210
211
211
212
212
V. CONCLUSIONS
It is expected that the accuracy of small field dosimetry
under a nonequilibrium condition can be significantly improved based on the following developments:
New protocols for absolute dosimetry in small and nonequilibrium condition will be developed to provide procedures for accurate absorbed dose calibrations for new
treatment modalities such as Gamma Knife, CyberKnife, tomotherapy, and other devices that do not
have standard reference field sizes. This will effectively
reduce the uncertainty and variations among different
centers in the absolute reference dose calibration.
Small volume detectors ion chambers, diodes, and others will be developed that have minimum perturbations
due to its presence and composition. Also, such detectors will have minimum energy, dose, and dose rate
dependence.
The accuracy of small field dosimetry will be greatly
improved by Monte Carlo simulations, especially under
extreme conditions, for small fields such as beam size
3 3 cm2 with inhomogeneity. Current measurement uncertainty 5% will be reduced. Monte Carlo
calculations will be able to accurately relate the dose to
a small field irradiated in an inhomogeneous media to
the dose of reference conditions in which the radiation
beam is calibrated. This will ensure that the uncertainty
of the dose determination in a small treatment field under extreme conditions remains similar in magnitude to
the uncertainty of a large reference field where the
beam is calibrated.
Monte Carlo simulations will provide correction parameters, such as correction factors for specific detectors
and for specific measurement conditions, and the stopping power ratio as a function of field size and beam
energy. These additional data will be available for routine use and will greatly reduce the measurement uncertainty under the nonequilibrium radiation conditions.
More accurate implementation of model-based calculation algorithms as well as direct Monte Carlo methods
will be available in commercial treatment planning systems for accurate dose calculation under the nonequilibrium radiation conditions.
213
213
tors using miniphantoms and brass caps, Med. Phys. 22, 11671170
1995.
24
I. A. E. A. Report No. 398, Absorbed Dose Determination in External
Beam Radiotherapy: An International Code of Practice for Dosimetry on
Standards of Absorbed Dose to Water, Technical Reports Series No. 398,
International Atomic Energy Agency, 2000.
25
Task Group-21, Radiation Therapy Committee, AAPM, A protocol for
the determination of absorbed dose from high-energy photon and electron
beams, Med. Phys. 10, 741771 1983.
26
T. Nyholm, J. Olofsson, A. Ahnesjo, and M. Karlsson, Modeling lateral
beam quality variations in pencil kernel based photon dose calculations,
Phys. Med. Biol. 51, 41114118 2006.
27
P. R. Almond, AAPMs TG-51 protocol for clinical reference dosimetry
of high-energy photon and electron beams, Med. Phys. 26, 18471870
1999.
28
P. Andreo and A. Brahme, Stopping power data for high-energy photon
beams, Phys. Med. Biol. 31, 839858 1986.
29
P. Andreo, J. R. Cunningham, K. Hohlfeld, and H. Svensson, Absorbed
Dose Determination in Photon and Electron Beams, An International
Code of Practice, Technical Report Series No. 277 International Atomic
Energy Agency, Vienna, 1987.
30
P. Andreo and A. E. Nahum, Stopping-power ratio for a photon spectrum
as a weighted sum of the values for monoenergetic photon beams, Phys.
Med. Biol. 30, 10551065 1985.
31
J. Seuntjens and F. Verhaegen, Comments on Ionization chamber dosimetry of small photon fields: A Monte Carlo study on stopping-power
ratios for radiosurgery and IMRT beams, Phys. Med. Biol. 48, L4345
2003.
32
S. Li, A. Rashid, S. He, and D. Djajaputra, A new approach in dose
measurement and error analysis for narrow photon beams beamlets
shaped by different multileaf collimators using a small detector, Med.
Phys. 31, 20202032 2004.
33
G. X. Ding, D. M. Duggan, B. Lu, D. E. Hallahan, A. Cmelak, A. Malcolm, J. Newton, D. Mathew, and C. W. Coffey, Impact of inhomogeneity corrections on dose coverage in treatment of lung cancer using
stereotactic body radiation therapy, Med. Phys. 34, 29852994 2007.
34
G. X. Ding, D. M. Duggan, and C. W. Coffey, Comment on Testing of
analytical anisotropic algorithm for photon dose calculation Med. Phys.
33, 41304148 2006, Med. Phys. 34, 3414 2007.
35
ICRU 31, Average energy required to produce an ion pair, ICRU Report 31, International Commission on Radiation Units and Measurements,
Washington, D.C., 1979.
36
R. Huber, D. Combecher, and G. Burger, Measurement of average energy required to produce an ion pair W value for low energy ions in
several gases, Radiat. Res. 101, 237251 1985.
37
I. J. Das, M. B. Downes, B. W. Corn, W. J. Curran, M. W. Wasik, and D.
W. Andrews, Characteristics of a dedicated linear accelerator-based stereotactic radiosurgery-radiotherapy unit, Radiother. Oncol. 38, 6168
1996.
38
S. C. Sharma, J. T. Ott, J. B. Williams, and D. Dickow, Commissioning
and acceptance testing of a CyberKnife linear accelerator, J. Appl. Clin.
Med. Phys. 8, 119125 2007.
39
A. Mack, S. G. Scheib, J. Major, S. Gianolini, G. Pazmandi, H. Feist, H.
Czempiel, and H. J. Kreiner, Precision dosimetry for narrow photon
beams used in radiosurgery-determination of Gamma Knife output factors, Med. Phys. 29, 20802089 2002.
40
P. Bjrk, T. Kns, and P. Nilsson, Measurements of output factors with
different detector types and Monte Carlo calculations of stopping-power
ratios for degraded electron beams, Phys. Med. Biol. 49, 44934506
2004.
41
A. L. McNiven, M. Mulligan, T. Kron, and J. J. Battista, The response of
prototype plane-parallel ionization chambers in small megavoltage x-ray
fields, Med. Phys. 33, 39974004 2006.
42
G. G. Zhang, D. W. O. Rogers, J. E. Cygler, and T. R. Mackie, Effects of
changes in stopping-power ratios with field size on electron beam relative
output factors, Med. Phys. 25, 17111716 1998.
43
H. Bouchard and J. Seuntjens, Ionization chamber-based reference dosimetry of intensity modulated radiation beams, Med. Phys. 31, 2454
2465 2004.
44
A. Wu, R. D. Zwicker, A. M. Kalend, and Z. Zheng, Comments on dose
measurements for a narrow beam in radiosurgery, Med. Phys. 20, 777
779 1993.
45
A. O. Jones, I. J. Das, and F. L. Jones, A Monte Carlo study of IMRT
214
70
214
A. S. Saini and T. C. Zhu, Dose rate and SDD dependence of commercially available diode detectors, Med. Phys. 31, 914924 2004.
71
TG-62, Diode In Vivo Dosimetry for Patients Receiving External Beam
Radiation Therapy: Report of the AAPM Radiation Therapy Committee
Task Group No. 62 Medical Physics Publishing, Madison, WI, 2005.
72
C. W. Cheng, S. H. Cho, M. Taylor, and I. J. Das, Determination of zero
field size percent depth doses and tissue maximum ratios for stereotactic
radiosurgery and IMRT dosimetry: Comparison between experimental
measurements and Monte Carlo simulation, Med. Phys. 34, 31493157
2007.
73
C. McKerracher and D. I. Thwaites, Assessment of new small-field detectors against standard-field detectors for practical stereotactic beam data
acquisition, Phys. Med. Biol. 44, 21432160 1999.
74
C. McKerracher and D. I. Thwaites, Notes on the construction of solidstate detectors, Radiother. Oncol. 79, 348351 2006.
75
P. Bjrk, T. Kns, and P. Nilsson, Comparative dosimetry of diode and
diamond detectors in electron beams for intraoperative radiation therapy,
Med. Phys. 27, 25802588 2000.
76
M. Heydarian, P. W. Hoban, W. A. Beckham, I. A. Borchardt, and A. H.
Beddoe, Evaluation of a PTW diamond detector for electron beam measurements, Phys. Med. Biol. 38, 10351042 1993.
77
P. W. Hoban, M. Heydarian, W. A. Beckham, and A. H. Beddoe, Dose
rate dependence of a PTW diamond detector in the dosimetry of a 6 MV
Photon beam, Phys. Med. Biol. 39, 12191229 1994.
78
V. S. Khrunov, S. S. Martynov, S. M. Vatnisky, I. A. Ermakov, A. M.
Chervjakov, D. L. Karlin, V. I. Fominych, and Y. V. Tarbeyev, Diamond
detectors in relative dosimetry of photon, electron and proton radiation
fields, Radiat. Prot. Dosimetry 33, 155157 1990.
79
W. U. Laub, T. W. Kaulich, and F. Nusslin, Energy and dose rate dependence of a diamond detector in the dosimetry of 425 MV photon
beams, Med. Phys. 24, 535536 1997.
80
S. Vatnitsky and H. Jrvinen, Application of natural diamond detector
for the measurement of relative dose distributions in radiotherapy, Phys.
Med. Biol. 38, 173184 1993.
81
Y. S. Horowitz, The theoretical and microdosimetric basis of thermoluminescence and applications to dosimetry, Phys. Med. Biol. 26, 765824
1981.
82
S. Pai, I. J. Das, J. F. Dempsey, K. L. Lam, T. J. LoSasso, A. J. Olch, J.
R. Palta, L. E. Reinstein, D. Ritt, and E. E. Wilcox, TG-69: Radiographic film for megavoltage beam dosimetry, Med. Phys. 34, 2228
2258 2007.
83
A. Niroomand-Rad, C. R. Blackwell, B. M. Coursey, K. P. Gall, J. M.
Galvin, W. L. McLaughlin, A. S. Meigooni, R. Nath, J. E. Rodgers, and
C. G. Soares, Radiographic film dosimetry: Recommendations of AAPM
Radiation Therapy Committee Task Group 55, Med. Phys. 25, 2093
2115 1998.
84
W. L. McLaughlin, C. G. Soares, J. A. Sayeg, E. C. McCullough, R. W.
Kline, A. Wu, and A. H. Maitz, The use of a radiochromic detector for
the determination of stereotactic radiosurgery dose characteristics, Med.
Phys. 21, 379388 1994.
85
R. Ramani, A. W. Lightstone, D. L. Mason, and P. F. OBrien, The use
of radiochromic film in treatment verification of dynamic stereotactic radiosurgery, Med. Phys. 21, 389392 1994.
86
R. Ramani, S. Russell, and P. F. OBrien, Clinical dosimetry using
MOSFETs, Int. J. Radiat. Oncol. Biol. Phys. 37, 959964 1997.
87
C. F. Chuang, L. Verhey, and P. Xia, Investigation of the use of
MOSFET for clinical IMRT dosimetric verification, Med. Phys. 29,
11091115 2002.
88
G. S. Ibbott, M. J. Maryanski, P. Eastman, S. D. Holcomb, Y. Zhang, R.
G. Avison, M. Sanders, and J. C. Gore, Three-dimensional visualization
and measurement of conformal dose distributions using magnetic resonance imaging of BANG polymer gel dosimeters, Int. J. Radiat. Oncol.
Biol. Phys. 38, 10971103 1997.
89
E. Pappas, L. Petrokokkinos, A. Angelopoulos, T. G. Maris, M. Kozicki,
I. Dalezios, and V. Kouloulias, Relative output factor measurements of a
5 mm diameter radiosurgical photon beam using polymer gel dosimetry,
Med. Phys. 32, 15131520 2005.
90
E. Pappas, T. G. Maris, A. Papadakis, F. Zacharopoulou, J. Damilakis, N.
Papanikolaou, and N. Gourtsoyiannis, Experimental determination of the
effect of detector size on profile measurements in narrow photon beams,
Med. Phys. 33, 37003710 2006.
91
H. Whitney, G. X. Ding, C. W. Coffey and J. C. Gore, Gel dosimetry for
measuring a complex 3D dose distribution in stereotactic radiosurgery,
215
103
215
P. Nizin, G. X. Qian, and H. Rashid, Zero-field dose data for 60C and
other high-energy photon beams in water, Med. Phys. 20, 13531360
1993.
104
P. S. Nizin and R. B. Mooij, An approximation of central-axis absorbed
dose in narrow photon beams, Med. Phys. 24, 17751780 1997.
105
P. S. Nizin and R. B. Mooij, Tissue-air ratios for narrow 60C gamma-ray
beams, Med. Phys. 25, 14581463 1998.
106
P. K. Kijewski, B. E. Bjarngard, and P. L. Petti, Monte Carlo calculations
of scatter dose for small field sizes in a 60C beam, Med. Phys. 13, 7477
1986.
107
G. X. Ding, D. M. Duggan, and C. W. Coffey, Commissioning stereotactic radiosurgery beams using both experimental and theoretical methods, Phys. Med. Biol. 51, 25492566 2006.
108
G. X. Ding, Dose discrepancies between Monte Carlo calculations and
measurements in the buildup region for a high-energy photon beam,
Med. Phys. 29, 24592463 2002.
109
G. X. Ding, Using Monte Carlo simulations to commission photon beam
output factors-A feasibility study, Phys. Med. Biol. 48, 38653874
2003.
110
A. Ahnesj, Collimator scatter in photon therapy beams, Med. Phys.
22, 267278 1995.
111
A. Mack, G. Mack, D. Weltz, S. G. Scheib, H. Bottcher, and V. Seifert,
High precision film dosimetry with GAFCHROMIC films for quality
assurance especially when using small fields, Med. Phys. 30, 23992409
2003.
112
M. Stasi, B. Baiotto, G. Barboni, and G. Scielzo, The behavior of several
microionization chambers in small intensity modulated radiotherapy
fields, Med. Phys. 31, 27922795 2004.
113
C. McKerracher and D. I. Thwaites, Verification of the dose to the isocentre in stereotactic plans, Radiother. Oncol. 64, 97107 2002.
114
F. Sanchez-Doblado, G. H. Hartmann, J. Pena, R. Capote, M. Paiusco, B.
Rhein, A. Leal, and J. I. Lagares, Uncertainty estimation in intensitymodulated radiotherapy absolute dosimetry verification, Int. J. Radiat.
Oncol. Biol. Phys. 68, 301310 2007.
system, radiation dose, interventional equipment and devices, procedure characteristics and treatment planning, and
personnel involved. We will first give an overview of the
current state of EIGI and then make predictions about each
feature category, providing the rationale and evidence for
each prediction and the strategy we suggest for making each
forecast become a reality in a 710 year time frame.
II. CURRENT STATUS
Before an EIGI procedure is scheduled, diagnostic procedures to find and analyze the pathological region are done
preferably using noninvasive imaging modalities, such as
multislice computed tomography MSCT or magnetic resonance imaging MRI, and followed up by higher-resolution
minimally invasive angiographic procedures done in dedicated angiographic or special procedure x-ray suites.5,6 Typically, EIGI procedures, if indicated, are then done under realtime x-ray image guidance in such a special procedure suite.7
Thus, such a suite may be used for the initial diagnostic
study and subsequently for the intervention. Rotating anode
x-ray tubes, that have become more robust but have changed
little in decades in overall design, are used as the source of x
rays; however, dynamic x-ray detector designs are in the process of changing from the decades old x-ray image intensifier
XII to the new flat panel detector FPD.8 The indirect FPD
and the XII designs both use an x-ray absorbing structured
CsI phosphor, but differ greatly thereafter. The XII converts
the phosphors light to electrons that are accelerated. This
electron distribution is focused on an output phosphor to
produce an intensified image of light photons and thus create
a gain in brightness.9 In contrast, an indirect FPD uses an
array of solid state photodiodes with thin film transistor
TFT switches and, currently, no additional gain to generate
0094-2405/2008/351/301/9/$23.00
301
302
302
III.A.1. Prediction 1
Higher resolution, lower noise, and real-time image
receptorsat first for the region of interest (ROI) near the
intervention and then across the full field of view (FOV)_
will become the standard of care for many EIGI procedures.
The rationale for this prediction is that, increasingly,
MSCT and MRI are taking over the task of vascular diagnosis with angiographic special procedure rooms being devoted
increasingly to EIGI. The fact that MSCT and magnetic resonance angiography MRA procedures require only a venous
303
303
resenting the signal through the TFT lines, where the noise is
added and then onto the sides of the sensor where it can be
amplified and digitized. Were this achievable in practice, the
problem for FPDs of reducing the size of the pixels would
still remain. Alternate detector designs actively being pursued by the authors consist of a high-sensitivity microangiographic fluoroscope HSMAF10 and a detector called the
solid state x-ray image intensifier SSXII. The HSMAF employs a light image intensifier between the CsI phosphor and
a digital videocamera with a fiber optic taper FOT input.
This detector has a small FOV sufficient to be used for EIGI
and is capable of real-time fluoroscopy as well as very-highresolution angiography. The SSXII consists of an array of
electron multiplying charge-coupled devices EMCCDs,43,44
each viewing a portion of the CsI phosphor through its own
FOT coupling.45 EMCCDs have all the advantages of standard CCDs: high speed, low noise, high resolution, and standard solid state production techniques. However, in addition,
they have a row of hundreds of multiplying elements each
with a small gain of a few percent. When cascaded together
in this row, they can result in a total gain from 1 to 2000
times, depending upon the variable gain voltage setting of
only around 20 V. The mosaic array of the SSXII also has the
advantage of being extensible in FOV by adding modules.
The INEE achievable for both such detectors is a fraction of
the exposure typical of low-dose fluoroscopy, hence they
have the desirable feature of being quantum limited throughout their range of operation. Additionally, unlike FPDs they
have no lag or ghosting and are available with small pixel
sizes. The potential advantages of SSXII and HSMAF detectors compared with conventional detectors are illustrated by
some examples in Figs. 1 and 2. A quantitative evaluation
using linear cascade analysis46 can be made of these new
high-resolution detectors45,47; however, recent additions to
these analytical methods will include the effects of other important factors that affect total system performance, such as
focal spot blurring and scatter in the patient.48,49
304
III.A.2. Prediction 2
High-resolution CBCT will become available for the ROI
of the interventional site with 3D roadmapping and automated indication of the catheter or guidewire tip superimposed.
The rationale for this prediction is that once a vascular
pathology has been diagnosed, superior image quality is
needed only over the site of the intervention and not over the
full volume FOV. Currently, there is some effort in both academic and commercial organizations to develop 3D roadmapping capability. The syngo iPilot product50 Siemens
Medical Solutions USA, Inc., Malvern, PA or the Dynamic
3D Roadmap product51 Philips Medical Systems, N.A,
Bothell, WA are such implementations with current detector
technology. Clearly, the higher the resolution available at the
interventional site, the greater the potential for a successful
intervention.
To achieve this goal, higher-resolution ROI detectors,
such as those in Prediction 1 above, mounted on stable detector holders added to angiographic CBCT-capable gantries
will be required. The associated software will also be
needed. Our group has shown that most of the artifacts due
to FOV truncation that one expects when the CT FOV is
smaller than the object being imaged can be eliminated by
combining the ROI data with a lower-.resolution full FOV
CT image sequence. This provides sufficient information
about those parts of the object that move in and out of the
ROI of the projection views but which are not actually located within the ROI or volume of interest of the 3D
object.52,53 These high-resolution ROI-CBCT images can
then be used as the basis for enhanced treatment planning by
calculating the simulation of the transport of a device
through the vasculature53,54 and for improved flow information, as will be discussed next.
III.A.3. Prediction 3
There will be greater accounting of patient dose distribution during EIGI with techniques to minimize integral dose
as well as deterministic effects.
Although patient dose for EIGI procedures can be some
of the largest of all medical procedures, it is highly nonuniform; hence, any determination of potential biological effects
will depend upon a detailed knowledge of the dose distribution. Although the European Union EU requires the measurement of dose-area product and the International Electrotechnical Commission Standard IEC 606012-43, 2000 and
the U.S. Food and Drug Administration FDA require that
fluoroscopic equipment include, during the procedure, the
display of air kerma rate and cumulative air kerma at an
interventional reference point.55,56 This is only a first step in
an accurate accounting of patient dose in EIGI. A past attempt to provide more detailed patient entrance surface dose
distributions in the form of a commercial product Caregraph, Siemens Medical Systems Corp., Malvern, PA was
apparently discontinued; however, as EIGI procedures become more complex, potentially longer, and with more nonMedical Physics, Vol. 35, No. 1, January 2008
304
III.B.1. Prediction 4
Endovascular devices will become finer, more patientspecific, more biocompatible, and more complex with the beginning of remotely actuated active components using shape
memory alloys (SMA), micromachines, microfluidics, and microelectronics; higher-resolution imaging will be relied upon
to assist in accurate device deployment.
Since minimally invasive EIGI procedures, even with the
current crop of simple and somewhat passive devices, are
rapidly replacing many invasive surgical procedures, it
would appear to make sense that this trend would continue
and be combined with advances in miniaturization. With
305
more accurate models of a specific patient vascular pathology obtained from CBCT, more patient-specific devices can
be created and deployed; however, care should be taken that
new endovascular devices are designed with constituent materials that are compatible with future imaging procedures
that might be required for the patient presently being treated,
so there is no negative effect such as artifact generation on
the images obtained.
As new more biocompatible materials are developed, devices such as bioabsorbable stents under investigation6366
should come into common usage. The initial development of
remote actuation is also happening. For example, external
magnetic fields are actively being used in the cardiovascular
area for accurately guiding new ablation probes for electrophysiology EP treatment,6769 and are being proposed in
the neurovascular arena for guiding the accurate deployment
of new flow diversion stents Fig. 1D for treatment of
aneurysms.70 The use of the existing magnetic field in MRI
systems is being reported for steering catheters.71 There is
the beginning commercial availability of remote catheter
steering systems, such as the one by Hansen Medical
Hansen Medical Inc., Mountain View, CA72,73 designed for
EP. However, these appear to be implemented with wires tied
to external motors and hence are hard to project to the full
range of EIGI, which may require navigation through a tortuous vessel. Other robotic systems have also been
reported.74 Remote actuation using inductive heating of
shape memory polymers is also being considered.75 There
are also a number of new more complex devices under development for the treatment of acute ischemic stroke, such as
ultrasonic clot busters, and complex catheter heads with high
speed saline streams to implement suction and hence reduce
clots.76 New technologies in radio-frequency ablation including software for image registration and fusion, thermal
modeling, electromagnetic tracking, semiautomated robotic
needle guidance, and multimodality imaging77 may also be
adaptable to EIGI.
To achieve the full potential for new patient-specific, biocompatible, active EIGI devices, basic components of the
process for creating these devices will have to be worked
out. First, improved morphological models of the patientspecific pathology will be achieved using the high-resolution
ROI-CBCT imaging methods described previously and using
computer models parameterizing the desired interventional
effect such as flow modification. See, for example, the CFD
calculations described next. An actual physical model of the
pathology might then be constructed using rapid-prototyping
techniques,78,79 and if the clinical circumstances permit, a
patient-specific device could be tested on the physical model
before actual clinical use. Next, if possible and if appropriate, a new biocompatible material will be selected so that the
device can be absorbed once the desired intervention has
been achieved, although reaching this goal may take longer
than the envisioned time period. Finally, the basic manufacturing process for generating the required functional elements of a new EIGI device will have to be further developed. For example, although it is doubtful that self-propelled
yet tethered devices will be commercialized in 710 years,
Medical Physics, Vol. 35, No. 1, January 2008
305
III.C.1. Prediction 5
Treatment planning for EIGI procedures will be more extensive and involve simulations to verify the optimal selection of devices and equipment for delivery as well as flow
calculations before, during, and after the intervention to
check the intervention as it proceeds.
The rationale for this prediction is that as more detailed
3D morphological information about a vascular pathology
becomes available from improved imaging studies and as
computer power continues to increase in availability at reduced cost, the use of simulations has begun83 and should
naturally continue to blossom. Evidence for this happening
in other areas, such as in radiation treatment planning, is
apparent. Patient-specific dose delivery regimens and resulting distributions are computer simulated with clinical CT
sequences as a basic part of the input data. For EIGI, however, there are two parts to a treatment plan that will have to
be simulated: The device delivery based upon vessel morphology and mechanical device properties, and the blood
flow results as determined using CFD calculations.
The implementation of increasingly sophisticated EIGI
treatment plans will probably occur gradually. Current research into simulations for device transport through tortuous
vasculature has begun; for example, with the tracking of a
rigid stent of given dimensions and determining tolerances
for passage along the parent vessel.53 Increasingly sophisticated models84 of the human vasculature, which include not
only the morphology as derived from CT sequences for pa-
306
306
the course of the EIGI to assist the interventionalist in making better decisions while the procedure is occurring. To further increase computer capability, it may be necessary to
have a fast digital link to a super-computer facility to enable
these complex calculations to be done so that actionable information is available during the EIGI.
III.D. Personnel involved
III.D.1. Prediction 6
FIG. 3. CFD result showing streamlines and local shear stress distributions
for a patient-specific aneurysm untreated and treated with an asymmetric
vascular stent Ref. 87. Notice effect of flow diversion on streamlines and
wall shear stress WSS distribution. A Streamlines, untreated; B streamlines, treated; C wall shear stress, untreated; D wall shear stress, treated.
The medical physicist will take a more active role in individual clinical EIGI procedures as well as in the training
of interventionalists using simulation systems.
If the future of EIGI procedures is as predicted herein,
with more patient intensive treatment planning, the analogy
of the medical physicists role to that in radiation therapy
treatment planning will be more plausible. As EIGI interventions become more technologically sophisticated, there will
be a need for a professional clinical physicist to help in the
selection of devices and delivery systems through simulations and quantitative analysis, as well as in overseeing the
appropriate interpretation and use of CFD results for individual patient interventions. These responsibilities, of course,
will be in addition to the primary continuing responsibility
for the specification and quality assurance of all imaging
systems.
Just as the therapy physicist is trained and certified in that
branch of medical physics, the vascular medical physicist
will have to be certified in EIGI physics as well. This implies
the necessity for new or augmented curricula in medical
physics training programs. While much of the basic hemodynamics and the CFD calculations may be presently in the
domain of biomechanical engineers, we expect that future
EIGI teams will consist of many medical professionals to
enable this rapidly advancing field to reach its vast potential.
However, it is our expectation that the vascular clinical
medical physicist will be uniquely qualified to integrate the
physics of imaging with the relevant aspects of biomechanical engineering involving interventional devices to be able to
oversee the technical aspects of EIGI facilities as well as to
head their scientific and engineering training programs for
medical personnel. Not only will there be a new syllabus of
EIGI physics related concepts to teach, but we expect that
simulation-based training of the physicians9499 who perform
EIGI procedures will become the standard. The interventionist will be able to practice and develop the necessary skills
for catheter navigation and device deployment in a programmable simulated environment with tactile and visual feedback. The EIGI medical physicist with the collaboration of
computer scientists will play a key role in both the use and
the development of such systems.
IV. CONCLUSIONS
In the 1966 movie Fantastic Voyage, there was an account
of what future endovascular brain surgery could be like. It
involved the shrinking of a submarine with a team of people
that was injected into a patient and instructed to take a route
through the vasculature to the brain to remove a blood clot in
307
307
pp. 231254.
S. Rudin, G. Yadava, G. Josan, A. Kuhls, H. Rangwala, Y. Wu, C. Ionita,
and D. R. Bednarek, New light-amplifier-based detector designs for high
spatial resolution and high sensitivity CBCT mammography, Proc. SPIE
6142, R1R11 2006.
11
P. R. Granfors, R. Aufrichtig, G. E. Possin, B. W. Giambattista, Z. S.
Huang, J. Liu, and B. Ma, Performance of a 41 41 cm2 amorphous
silicon flat panel x-ray detector designed for angiographic and R&F imaging applications, Med. Phys. 30, 27152726 2003.
12
A. K. Bloomquist, M. J. Yaffe, G. E. Mawdsley, D. M. Hunter, and D. J.
Beideck, Lag and ghosting in a clinical flat-panel selenium digital mammography system, Med. Phys. 33, 29983005 2006.
13
J. H. Siewerdsen and D. A. Jaffray, A ghost story: Spatio-temporal response characteristics of an indirect-detection flat-panel imager, Med.
Phys. 26, 16241641 1999.
14
C. Schmidgunst, D. Ritter, and E. Lang, Calibration model of a dual gain
flat panel detector for 2D and 3D x-ray imaging, Med. Phys. 34, 3649
3666 2007.
15
A. Ganguly, S. Rudin, D. R. Bednarek, K. R. Hoffmann, and I. Kyprianou, Micro-angiography for neuro vascular imaging, Part 1: Experimental measurements and feasibility, Med. Phys. 30, 30183028 2003.
16
Food and Drug Administration, FDA Public Health Advisory, Avoidance
of serious x-ray induced skin injuries to patients during fluoroscopicallyguided procedures, September 30, 1994, Center for Devices and Radiological Health, Rockville, Maryland available at http://www.fda.gov/
cdrh/fluor.html.
17
L. K. Wagner, M. D. McNeese, M. V. Marx, and E. L. Siegel, Severe
skin reactions from interventional fluoroscopy: Case report and review of
the literature, Radiology 213, 773776 1999.
18
S. Rudin and D. R. Bednarek, Region of interest fluoroscopy, Med.
Phys. 19, 11831189 1992.
19
S. Rudin, D. R. Bednarek, L. R. Guterman, A. Wakhloo, L. N. Hopkins,
L. Fletcher, and P. Massoumzadeh, Implementation of region of interest
fluoroscopy using the road mapping mode of a real-time digital radiographic unit, Radiographics 15, 14651470 1995.
20
S. Rudin, L. R. Guterman, W. Granger, D. R. Bednarek, and L. N. Hopkins, Neuro-interventional radiologic application of region of interest
ROI imaging techniques, Radiology 199, 870873 1996.
21
S. Rudin, D. R. Bednarek, M. Kezerashvili, W. Granger, J. Serghany, L.
R. Guterman, L. N. Hopkins, P. Massoumzadeh, B. Szymanski, and R.
Loftus, Clinical application of region of interest ROI radiologic imaging techniques, Radiographics 16, 895902 1996.
22
N. A. Gkanatsios, W. Huda, K. R. Peters, and J. A. Freeman, Evaluation
of an on-line patient exposure meter in neuroradiology, Radiology 203,
837842 1997.
23
S. Balter, D. W. Fletcher, H. M. Kuan, D. Miller, D. Richter, H. Seissl,
and T. B. Shope, Techniques to estimate radiation dose to skin during
fluoroscopically guided procedures, in Intravascular Brachytherapy/
Fluoroscopically-Guided Interventions, edited by S. Balter, R. Chan, and
T. Shope AAPM Summer School Proceedings, Madison, Wisconsin,
2002, pp. 731764.
24
C. T. Dotter and M. P. Judkins, Transluminal treatment of arteriosclerotic
obstruction. Description of a new technic and a preliminary report of its
application, 1964, Radiology 172, 904920 1989 classical article.
25
H. Ceschinski, H. Henkes, H. C. Weinert, W. Weber, D. Kuhne, and H.
Monstadt, Torquability of microcatheter guidewires: the resulting torsional moment, Biomed. Mater. Eng. 10, 3142 2000.
26
J. Carey, A. Fahim, and M. Munro, Design of braided composite and
cardiovascular catheters based on required axial, flexural and torsional
rigidities, J. Biomed. Mater. Res., Part B: Appl. Biomater. 70B, 7381
2004.
27
J. Carey, D. Emery, and P. McCracken, Buckling test as a new approach
to testing flexural rigidities of angiographic catheters, J. Biomed. Mater.
Res., Part B: Appl. Biomater. 76B, 211218 2006.
28
P. Bloss, W. Rothe, P. Wunsche, C. Werner, A. Rothe, G. D. Kneissl, W.
Burger, and E. Rehberg, Investigations of the pushability behavior of
cardiovascular angiographic catheters, Biomed. Mater. Eng. 13, 327343
2003.
29
G. H. Zoarski, J. M. Mathis, and J. R. Hebel, Performance characteristics
of microcatheter systems in a standard tortuous pathway, AJNR Am. J.
Neuroradiol. 19, 15711576 1998.
30
J. M. Anderson, The future of biomedical materials, J. Mater. Sci.
Mater. Med. 17, 10258 2006 review.
10
308
31
308
view FFOV and truncated CT for a human head phantom, Proc. SPIE
5745, 583590 2005.
53
K. R. Hoffmann, S. Rudin, H. Meng, L. N. Hopkins, L. Guterman, and E.
Levy, in Multidimensional Image Processing, Analysis, and Display, edited by S. G. Armato and M. S. Brown RSNA, Oak Brook, Illinois,
2005, pp. 173184.
54
S. Schafer, V. Singh, K. R. Hoffmann, P. B. Nol, and J. Xu, Planning
image-guided endovascular interventions: guidewire simulation using
shortest path algorithms, Proc. SPIE 6509, C1-C10 2007.
55
International Electrotechnical Commission, Report 606012-43, Medical
electrical equipment Part 2-43: Particular requirements for the safety of
x-ray equipment for interventional procedures, Geneva, Switzerland,
2000.
56
U. S. Food and Drug Administration FDA, HHS: Electronic Products
Federal performance standard for diagnostic x-ray systems and their major components, Federal Register, June 10, 2005 Vol. 70, Number 111,
pp. 3399734042.
57
P. Massoumzadeh, S. Rudin, and D. R. Bednarek, Filter material selection for region of interest radiologic imaging, Med. Phys. 25, 161171
1998.
58
P. M. Dinu, D. R. Bednarek, D. Wobschall, R. Peterson, C. N. Ionita, S.
Rudin, M. Zeng, K. Chugh, and K. R. Hoffmann, X-ray beam tracking
system for a fluoroscopic c-arm unit, Med. Phys. 30, 1424 2003.
59
K. Chugh, P. M. Dinu, D. R. Bednarek, D. Wobschall, S. Rudin, K. R.
Hoffmann, R. Peterson, and M. Zeng, A computer-graphic display for
real-time operator feedback during interventional x-ray procedures,
Proc. SPIE 5367, 464473 2004.
60
Food and Drug Administration, Important Information for Physicians and
other Health Care Professionals, Recording information in the patients
medical record that identifies the potential for serious x-ray-induced skin
injuries following fluoroscopically-guided procedures, September 15,
1995, Center for Devices and Radiological Health, Rockville Maryland
available at http://www.fda.gov/cdrh/xrayinj.html.
61
D. L. Miller, S. Balter, L. K. Wagner, J. Cardella, T. W. Clark, C. D.
Neithamer, Jr., M. S. Schwartzberg, T. L. Swan, R. B. Tobwin, K. S.
Rholl, and D. Sacks, SIR standards of Practice Committee, Quality
improvement guidelines for recording patient radiation dose in the medical record, J. Vasc. Interv. Radiol. 15, 423429 2004.
62
L. K. Wagner, in Intravascular Brachytherapy / Fluoroscopically-Guided
Interventions, edited by S. Balter, R. Chan, and T. Shope AAPM Summer School Proceedings, Madison, Wisconsin, 2002, pp. 195218.
63
J. A. Ormiston, M. W. I. Webster, and G. Armstrong, First-in-human
implantation of a fully bioabsorbable drug-eluting stent: the BVS poly-Llactic acid everolimus eluting coronary stent, Catheter. Cardiovasc. Interv. 69, 128131 2007.
64
R. Erbel et al., Temporary scaffolding of coronary arteries with bioabsorbable magnesium stents: a prospective, non-randomised multicentre
trial, Lancet 369, 18691875 2007.
65
J. Ormiston and M. Webster, Absorbable coronary stents. comment,
Lancet 369, 18391840 2007.
66
S. Saito, New horizon of bioabsorbable stent. comment, Catheter.
Cardiovasc. Interv. 66, 595596 2005 editorial.
67
http://www.stereotaxis.com/
68
M. P. Armacost, J. Adair, T. Munger, R. Viswanathan, F. M. Creighton,
D. T. Curd, and R. Sehra, Accurate and reproducible target navigation
with the stereotaxis Niobe Magnetic Navigation System, J. Cardiovasc.
Electrophysiol. 18, S26S31 2007.
69
C. Pappone et al., Robotic magnetic navigation for atrial fibrillation
ablation, J. Am. Coll. Cardiol. 47, 13901400 2006.
70
E. Florin, H. S. Rangwala, and S. Rudin, Method to rotate an endovascular device around the axis of a vessel using an external magnetic field,
Med. Phys. 34, 328333 2007.
71
F. Settecase, M. S. Sussman, M. W. Wilson, S. Hetts, R. L. Arenson, V.
Malba, A. F. Bernhardt, W. Kucharczyk, and T. P. L. Roberts,
Magnetically-assisted remote control MARC steering of endovascular
catheters for interventional MRI: A model for deflection and design implications, Med. Phys. 34, 31353142 2007.
72
http://www.hansenmedical.com/products/sensei.aspx
73
A. D. Al-Ahmad, J. Grossman, and P. J. Wang, Early experience with a
computerized robotically controlled catheter system, J. Interv. Card.
Electrophysiol. 12, 199202 2005.
74
L. Cercenelli, E. Marcelli, and G. Plicchi, Initial experience with a
telerobotic system to remotely navigate and automatically reposition stan-
309
309
namic CFD calculations in the computed tomography CT derived lumen, Proc. SPIE 6143, G1G8 2006.
88
J. R. Cebral, R. S. Pergolizzi, Jr., and C. M. Putnam, Computational fluid
dynamics model of intracranial aneurysms: qualitative comparison with
cerebral angiography, Acad. Radiol. 14, 804813 2007.
89
J. Cebral, A. Radaelli, A. Frangi, and C. Putman, Hemodynamics before
and after bleb formation in cerebral aneurysms, Proc. SPIE 6511,
65112C165112C9 2007.
90
S. Appanaboyina, F. Mut, R. Lhner, C. Putman, and J. Cebral, Patient
specific modeling of intracranial aneurysmal stenting, Proc. SPIE 6511,
65112D165112D11 2007.
91
Z. Wang, C. N. Ionita, S. Rudin, K. R. Hoffmann, A. B. Paxton, and D. R.
Bednarek, Angiographic analysis of blood flow modification in cerebral
aneurysm models with a new asymmetric stent, Proc. SPIE 5369, 307
318 2004.
92
S. Rudin, Z. Wang, I. Kyprianou, K. R. Hoffmann, Y. Wu, H. Meng, L. R.
Guterman, B. Nemes, D. R. Bednarek, J. Dmochowski, and L. N. Hopkins, Measurement of flow modification in phantom aneurysm model:
comparison of coils and a longitudinally and axially asymmetric stent
initial findings, Radiology 231, 272276 2004.
93
B. B. Lieber, C. Sadasivan, L. Miskolczi, L. Cesar, J. Seong, and A. K.
Wakhloo, Flow divertors to treat cerebral aneurysms: preliminary results
in the rabbit elastase-induced aneurysm model, in Proceedings BIO2006,
ASME 2006 Summer Bioengineering Conference, June 2125, 2006,
Amelia Island Plantation, Amelia Island, Florida.
94
T. Alderliesten, M. K. Konings, and W. J. Niessen, Simulation of minimally invasive vascular interventions for training purposes, Comput.
Aided Surg. 9, 315 2004.
95
T. Alderliesten, M. K. Konings, and W. J. Niessen, Modeling friction,
intrinsic curvature, and rotation of guidewires for simulation of minimally
invasive vascular interventions, IEEE Trans. Biomed. Eng. 54, 2939
2007.
96
C. Duriez, S. Cotin, J. Lenoir, and P. Neumann, New approaches to
catheter navigation for interventional radiology simulation, Comput.
Aided Surg. 11, 300308 2006.
97
D. L. Dawson, Training in carotid artery stenting: Do carotid simulation
systems really help?, Vascular 14, 256263 2006 review.
98
D. A. Gould, D. O. Kessel, A. E. Healey, S. J. Johnson, and W. E.
Lewandowski, Simulators in catheter-based interventional radiology:
training or computer games?, Clin Radiol. 61, 556561 2006 review,
D. A. Gould, D. O. Kessel, A. E. Healey, S. J. Johnson, and W. E.
Lewandowski, Clin. Radiol. 61, 903 2006 erratum.
99
A. Sengupta, T. Kesavadas, K. R. Hoffmann, R. E. Baier, and S. Schafer,
Evaluating tool-artery interaction force during endovascular neurosurgery for developing haptic engine, Stud. Health Technol. Inform. 125,
418420 2007.
Bruno De Manc
CT and X-ray Laboratory, GE Global Research, Niskayuna, New York 12309
Received 5 October 2007; revised 20 December 2007; accepted for publication 20 December 2007;
published 25 February 2008
Over the past decade, computed tomography CT theory, techniques and applications have undergone a rapid development. Since CT is so practical and useful, undoubtedly CT technology will
continue advancing biomedical and non-biomedical applications. In this outlook article, we share
our opinions on the research and development in this field, emphasizing 12 topics we expect to be
critical in the next decade: analytic reconstruction, iterative reconstruction, local/interior reconstruction, flat-panel based CT, dual-source CT, multi-source CT, novel scanning modes, energy-sensitive
CT, nano-CT, artifact reduction, modality fusion, and phase-contrast CT. We also sketch several
representative biomedical applications. 2008 American Association of Physicists in Medicine.
DOI: 10.1118/1.2836950
Key words: analytic reconstruction, iterative reconstruction, local/interior reconstruction, flat-panel
based CT, dual-source CT, multi-source CT, scanning mode, energy-sensitive CT, nano-CT, artifact
reduction, modality fusion, phase-contrast CT
I. INTRODUCTION
The evolution of civilization has been largely driven by the
need for extending humans capabilities. Arguably, the most
important way for us to sense the world is by visual perception. However, the human vision is severely limited by the
opaqueness of many natural and artificial objects. Hence,
how to achieve an inner vision was often pondered in various scenarios through history. Mainly credited to the pioneering works of Cormack1 and Hounsfield,2 in the last century, x-ray computed tomography CT is the first imaging
modality that allows accurate non-destructive interior image
reconstruction of an object from a sufficient number of x-ray
projections. Hounsfields x-ray CT prototype immediately
generated a tremendous excitement in the medical community and inspired a rapid technical development with an ever
strong momentum. Also, x-ray CT as the first trans-axial tomography model promoted the development of other tomographic modalities for biomedical applications and beyond,
such as magnetic resonance imaging, ultrasound tomography,
nuclear tomography, optical tomography, molecular tomography, and so on.
Since its introduction in 1973,2 x-ray CT has revolutionized radiographic imaging and become a cornerstone of every modern radiology department. Closely correlated to the
development of x-ray CT, the research for higher performance system architectures and more advanced image reconstruction algorithms has been intensively pursued for important biomedical applications. A famous CT scanner is the
dynamic spatial reconstructor DSR built at the Mayo Clinic
in 1979.3,4 With the ability to acquire 240 contiguous 1 mm
slices in a time window of 0.01 s, the heart, lung, and blood
flow can be vividly observed. This DSR system is considered
as the precursor of the electron-beam CT scanner, the dual1051
0094-2405/2008/353/1051/14/$23.00
1051
1052
1052
TABLE I. Representative search data on CT studies and application, where
the percentage means the number of hits for the past five years over that for
the past ten years searched on 9/4/07.
Rule
CT
CT
CT
CT
CT
CT
CT
CT
CT
CT
CT
and
and
and
and
and
and
and
and
and
and
and
algorithm
cancer
cardiac
dose
dynamic
dual-energy
screen
therap
diagno
fusion
mouse
No. of Hits
for 97-07
No. of Hits
for 02-07
2785
11 468
2252
5595
2572
235
2771
12 099
27 025
1586
787
1829
7642
1517
3609
1456
142
1812
7296
15604
1134
416
65.7
66.6
67.4
64.5
56.7
60.4
65.4
60.3
57.7
71.5
52.9
Since Katsevichs 2002 paper on exact and efficient helical cone-beam CT reconstruction,14 intensified research efforts have been made in this area.15 Various sophisticated
formulas have been proposed for exact reconstruction from
projection data that can be longitudinally truncated and even
transversely truncated, and for not only a standard helical
trajectory but also a quite general class of scanning loci Fig.
2.1624 However, in the general case such as saddle-like
scanning paths24 and nonstandard helical trajectories, these
algorithms are far less computationally efficient as compared
to the popular filtered backprojection FBP with spatially
invariant filtering. In the 9th International Meeting on Fully
3D Image Reconstruction in Radiology and Nuclear Medicine Lindau, Germany, July 913, 2007, Katsevich presented an important progress towards exact yet efficient general cone-beam reconstruction algorithms for two classes of
scanning loci.25 The first class curves are smooth and of positive curvature and torsion. The second class consists of
circle-plus curves, with the segment for the plus part starting
below the circle-like trajectory and ending above it.26 However, there are other important classes of trajectories for
which exact and efficient algorithms are desired and highly
nontrivial. Therefore, it remains an open challenge to formulate such exact and efficient algorithms for many other types
of scanning curves. Theoretical unification of these new exact algorithms is also worthwhile.2729
TABLE II. Categorization of 262 relevant papers selected from the 363 articles in English retrieved under the rule x-ray and CT for 2007
searched on 9/4/07.
Category
No. of Papers
72
127
14
49
27.5
48.5
5.3
18.7
1053
FIG. 2. Representative general scanning trajectories. a A non-standard helix, b a saddle curve and c a circle-plus-line combination.
1053
There are two reasons. First, in a good number of applications, the data completeness required for exact reconstruction
cannot be satisfied due to physical constraints. Hence, the
only choice is to perform an approximate reconstruction.
Second, even if an exact reconstruction is practically feasible, oftentimes the approximate algorithms can deliver
similar or even better performance as compared to the exact
reconstruction counterparts, which are not necessarily optimal in terms of the noise characteristics. Figure 3 shows two
approximate reconstructions of excellent quality.33
It is our opinion that the popularity of approximate conebeam reconstruction algorithms will definitely sustain in the
near future. With the development of exact cone-beam reconstruction algorithms, the approximate cone-beam reconstruction algorithms will be surely improved as well. In addition
to various approximations to be improved or made on an
individual basis, a promising research direction for approximate cone-beam reconstruction would be to adapt new exact
cone-beam reconstruction algorithms.3436 By doing so, the
merits of exact cone-beam reconstruction algorithms can be
largely inherited at a much lower computational cost and
modified to have certain practically desirable features. This
approach should be particularly attractive in the cases of incomplete and/or inconsistent data such as cardiac CT, contrast studies, artifact reduction, and so on.
III.B. Iterative reconstruction
1054
1054
FIG. 4. Images analytically and iteratively reconstructed from clinical data. a A FBP reconstruction from a 300 mAs dataset, b a FBP reconstruction from
a 75 mAs dataset, and c an iterative reconstruction from the same 75 mAs dataset Ref. 48 Courtesy of J.-B. Thibault.
noisy. Even in the cases where analytic reconstruction performs well, there is no fundamental reason why iterative reconstruction would perform any worse. Hence, we predict
that the fast development in computing methods and hardware will lead to a shift from analytic to statistical reconstruction or at least a fusion of analytic and iterative approaches. This evolution will happen gradually, because of
the high computational cost as well as the time needed for
the radiologists to adopt this new technology.
III.C. Local/interior CT
1055
1055
1056
1056
1057
1057
FIG. 7. Novel source configurations. a A distributed source with multiple spots along the z direction to eliminate cone-beam artifacts Ref. 80, and b an
inverse-geometry architecture with tens of spots combining the benefits of a line source in z, a small photon-counting detector, and a virtual bowtie Ref. 83.
Figure 7a is duplicated from Ref. 80 with the permission of GE Company.
FIG. 8. Compositing-circling scanning mode. In such a CT system, the scanning trajectory is a composition of two circular motions: While an x-ray
focal spot is rotated on a plane facing an object to be reconstructed, the
x-ray source is also rotated around the object on the gantry plane. Once a
projection dataset is acquired, exact or approximate reconstruction can be
done Copyright by G. Wang and H. Y. Yu, US Provisional Patent Application, 2007.
1058
1058
1059
1059
FIG. 10. Representative CT image artifacts. a A shoulder phantom image with streaks caused by photon starvation, b a patient with spine implants
generating metal artifacts, and c a moving head leading to motion artifacts Duplicated from Ref. 100 with the permission of the Radiological Society of
North America.
1060
1060
FIG. 11. Imaging platform Inveon for fusion of CT, PET and SPECT in
preclinical applications Courtesy of Siemens Company.
explicit relationships describing dependence among the involved datasets. Such an integrated inverse problem may require an iterative solution containing several loops each of
which assumes other image reconstructions known and refines an intermediate image, or have more sophisticated
forms like a truly simultaneous iterative solution.114 Theoretical studies are needed to establish the solution existence,
uniqueness and stability with new iterative reconstruction
schemes, as already mentioned for Topics B and J. In the
cases of no unique solutions, regularization issues must be
addressed with the aid of a priori knowledge in the form of
constraints, penalty terms and so on.
III.L. Phase-contrast CT
In all mainstream x-ray CT imaging modalities, the contrast mechanism has been attenuation based for over a century. As a result, weak-absorbing tissues are not imaged well,
and radiation dose has been a general concern. On the other
hand, x-ray phase-contrast imaging relies on the diffraction
properties of structures, promising to have much better contrast at much lower dose. Specifically, for x-rays the refraction index of biological soft tissues is approximately 1.0,
namely, n = 1 + i, where and represent attenuation
and diffraction, respectively. In the range of 15 150 KeV,
is about three orders of magnitude greater than , making
phase-contrast imaging about three orders of magnitude
more sensitive than attenuation-based methods.115 Typical
values of refraction indexes of breast tissue are plotted in
Ref. 116.
Medical Physics, Vol. 35, No. 3, March 2008
1061
1061
cone-beam technologies has increased and continues increasing the usage of CT. There is a substantial room for dose
reduction using algorithmic means for attenuation-based CT.
Other possible directions include more dose-efficient scanning protocols, better a priori knowledge, more effective reconstruction methods and smarter post-processing strategies.
Phase-contrast based CT, if successfully developed and clinically utilized, will revolutionize its x-ray imaging applications by virtually eliminating the dose problems and greatly
boosting contrast resolution, which may open doors to novel
functional and molecular imaging studies, and overtake or
complement some MRI applications. X-ray phase-contrast
CT/micro-CT is a technically very challenging and relatively
immature area, but it has the potential to have huge impact in
certain pre-clinical and clinical applications.
As far as temporal resolution is concerned, cardiac CT is
by far the most prominent application.131 Most common CT
targets are relatively motionless, such as bony structures,
head, neck, and abdomen. However, the closer we get to the
heart, the poorer the image quality becomes, because of the
cardiac motion. The coronary arteries are too elusive to be
clearly captured in challenging cases such as when the cardiac rate is too high or irregular, since the relative rest phase
of the coronary arteries is only about 60 ms. Most demanding cardiac imaging used to be done by EBCT, which is
expensive and rarely available. In addition to its remarkable
applications for dynamic anatomical imaging of cardiac
structures, EBCT is also a powerful tool for physiological
imaging. Following the introduction of the Siemens dualsource scanner, it seems that more efforts are warranted to
design better multi-source or distributed source systems and
methods for cardiac CT. The ideas and schemes already exist
on triple-source and multi-source CT systems, and hopefully
will be refined and realized in the not too distant future.
Finally, CT images with a high quantitative accuracy will
be more informative in general and extremely useful with
energy-sensitive CT in particular. It is anticipated that more
advanced x-ray sources such as portable synchrotron radiation devices will be much more accessible one or two decades from now. In the meantime, innovative sources and
detectors will be developed to generate spectrally resolved
projection data directly or indirectly. Statistical reconstruction can be applied to design and refine iterative algorithms
that fully utilize all detected photons, and add a spectral dimension to CT images. Presumably, this type of energysensitive CT will dramatically improve the characterization
of heart and lung diseases as well as various cancers. Along
this direction, scattering-based CT, x-ray induced fluorescence CT, and CT in other hybrid modes may also become
useful.
V. CONCLUDING REMARKS
When the first author of this paper graduated with a CT
dissertation in 1992, he considered to switch to a different
imaging field. His former mentor, Dr. Vannier, who is an
authority in x-ray imaging and a certified radiologist, advised
him not to do so because CT was just too practical and
1062
1062
cone-beam spiral/helical CT, IEEE Trans. Med. Imaging 199, 817821
2000.
12
L. A. Feldkamp, L. C. Davis, and J. W. Kress, Practical cone-beam
algorithm, J. Opt. Soc. Am. A 1A, 612619 1984.
13
A. Katsevich, An improved exact filtered backprojection algorithm for
spiral computed tomography, Adv. Appl. Math. 324, 681697 2004.
14
A. Katsevich, Theoretically exact filtered backprojection-type inversion
algorithm for spiral CT, SIAM J. Appl. Math. 626, 20122026 2002.
15
G. Wang, Y. B. Ye, and H. Y. Yu, Appropriate and exact cone-beam
reconstruction with standard and nonstandard spiral scanning, Phys.
Med. Biol. 526, R1R13 2007.
16
Y. B. Ye, S. Y. Zhao, H. Y. Yu, and G. Wang, Exact reconstruction for
cone-beam scanning along nonstandard spirals and other curves, Proceedings of SPIE, Vol. 5535, pp. 293300, Denver, CO 2004.
17
Y. B. Ye and G. Wang, Filtered backprojection formula for exact image
reconstruction from cone-beam data along a general scanning curve,
Med. Phys. 321, 4248 2005.
18
Y. B. Ye, S. Y. Zhao, H. Y. Yu, and G. Wang, A general exact reconstruction for cone-beam CT via backprojection-filtration, IEEE Trans.
Med. Imaging 249, 11901198 2005.
19
J. Pack and F. Noo, Cone-beam reconstruction using 1D filtering along
the projection of M-lines, Inverse Probl. 214, 11051120 2005.
20
J. D Pack, F. Noo, and R. Clackdoyle, Cone-beam reconstruction using
the backprojection of locally filtered projections, IEEE Trans. Med. Imaging 241, 7085 2005.
21
T. L. Zhuang, S. Leng, B. E. Nett, and G. H. Chen, Fan-beam and
cone-beam image reconstruction via filtering the backprojection image of
differentiated projection data, Phys. Med. Biol. 4924, 54895503
2004.
22
Y. Zou, X. Pan, and E. Y. Sidky, Theory and algorithms for image
reconstruction on chords and within regions of interest, J. Opt. Soc. Am.
A 2211, 23722384 2005.
23
G. Wang and Y. B. Ye, Nonstandard spiral cone-beam scanning methods,
apparatus, and applications, US Provisional Patent Application 60/
588,682, Filing date: July 16, 2004 2004.
24
G. Wang and H. Y. Yu, Scanning method and system for cardiac imaging, Patent disclosure submitted to Virginia Tech. Intellectual Properties
on July 17 2007.
25
A. Katsevich and M. Kapralov, Theoretically exact FBP reconstruction
algorithms for two general classes of curves, 9th International Meeting
on Fully Three-Dimensional Image Reconstruction in Radiology and
Nuclear Medicine, pp. 8083, Lindau, Germany 2007.
26
A. Katsevich, Image reconstruction for a general circle-plus trajectory,
Inverse Probl. 235, 22232230 2007.
27
A. Katsevich, A general scheme for constructing inversion algorithms
for cone-beam CT, Int. J. Math. Math. Sci. 21, 13051321 2003.
28
Y. C. Wei, H. Y. Yu, J. Hsieh, W. X. Cong, and G. Wang, Scheme of
computed tomography, J. X-Ray Sci. Technol. 154, 235270 2007.
29
S. Y. Zhao, H. Y. Yu, and G. Wang, A unified framework for exact
cone-beam reconstruction formulas, Med. Phys. 326, 17121721
2005.
30
G. H. Chen, An alternative derivation of Katsevichs cone-beam reconstruction formula, Med. Phys. 3012, 32173226 2003.
31
H. Y. Yu and G. Wang, Cone-beam composite-circling scan and exact
image reconstruction for a quasi-short object, Int. J. Biomed. Imaging
2007, Article ID 87319, 10 pages 2007.
32
J. D. Pack, F. Noo, and H. Kudo, Investigation of saddle trajectories for
cardiac CT imaging in cone-beam geometry, Phys. Med. Biol. 4911,
23172336 2004.
33
X. Y. Tang, J. Hsieh, R. A. Nilsen, S. Dutta, D. Samsonov, and A. Hagiwara, A three-dimensional-weighted cone beam filtered backprojection
CB-FBP algorithm for image reconstruction in volumetric CT - helical
scanning, Phys. Med. Biol. 514, 855874 2006.
34
X. Y. Tang and J. Hsieh, A filtered backprojection algorithm for cone
beam reconstruction using rotational filtering under helical source trajectory, Med. Phys. 3111, 29492960 2004.
35
X. Y. Tang and J. Hsieh, Handling data redundancy in helical cone beam
reconstruction with a cone-angle-based window function and its
asymptotic approximation, Med. Phys. 346, 19891998 2007.
36
X. Y. Tang, J. Hsieh, R. A. Nilsen, and S. M. McOlash, Extending
three-dimensional weighted cone beam filtered backprojection CB-FBP
algorithm for image reconstruction in volumetric CT at low helical
pitches, Int. J. Biomed. Imaging 2006, Article ID 45942, 8 pages 2006.
1063
37
R. Gordon, R. Bender, and G. T. Herman, Algebraic reconstruction techniques ART for three-dimensional electron microscopy and x-ray photography, J. Theor. Biol. 29, 471482 1970.
38
A. H. Andersen and A. C. Kak, Simultaneous algebraic reconstruction
technique SART: a superior implementation of the ART algorithm,
Ultrason. Imaging 6, 8194 1984.
39
L. A. Shepp and B. F. Logan, The Fourier reconstruction of a head
section, IEEE Trans. Nucl. Sci. NS213, 2134 1974.
40
A. J. Rockmore and A. Macovski, A maximum likelihood approach to
emission image reconstruction from projections, IEEE Trans. Nucl. Sci.
234, 14281432 1976.
41
J. Nuyts et al., Iterative reconstruction for helical CT: A simulation
study, Phys. Med. Biol. 434, 729737 1998.
42
B. De Man et al., Reduction of metal streak artifacts in x-ray computed
tomography using a transmission maximum a posteriori algorithm,
IEEE Trans. Nucl. Sci. 473, 977981 2000.
43
I. A. Elbakri and J. A. Fessler, Statistical image reconstruction for polyenergetic x-ray computed tomography, IEEE Trans. Med. Imaging 212,
8999 2002.
44
J. A. Fessler, Statistical image reconstruction methods for transmission
tomography, in Medical Image Processing and Analysis, edited by M.
Sonka and J. M. Fitzpatric SPIE Press, Bellingham, Washington, 2000,
Vol. 3, pp. 170.
45
J. A. Fessler and A. O. Hero, Space-alternating generalized expectationmaximization algorithm, IEEE Trans. Signal Process. 4210, 2664
2677 1994.
46
I. A. Elbakri and J. A. Fessler, Segmentation-free statistical image reconstruction for polyenergetic x-ray computed tomography with experimental validation, Phys. Med. Biol. 4815, 24532477 2003.
47
P. E. Kinahan, A. M. Alessio, and J. A. Fessler, Dual energy CT attenuation correction methods for quantitative assessment of response to cancer therapy with PET/CT imaging, Technol. Cancer Res. Treat. 54,
319327 2006.
48
J. B. Thibault, A. K. Sauer, A. C. Bouman, and J. Hsieh, Threedimensional statistical modeling for image quality improvements in
multi-slice helical CT, 8th Meeting on Fully ThreeDimensional Image
Reconstruction in Radiology and Nuclear Medicine, pp. 271274, Salt
Lake City, UT 2005.
49
M. Iatrou, B. De Man, and S. Basu, A comparison between filtered
backprojection, post-smoothed weighted least squares, and penalized
weighted least squares for CT reconstruction, IEEE Nuclear Science
Symposium Conference Record, 2006, pp. 28452850, San Diego, CA
2006.
50
D. L. Parker, Optimal short scan convolution reconstruction for fanbeam
CT, Med. Phys. 92, 254257 1982.
51
Y. Zou and X. C. Pan, Exact image reconstruction on PI-lines from
minimum data in helical cone-beam CT, Phys. Med. Biol. 496, 941
959 2004.
52
M. Defrise, F. Noo, R. Clackdoyle, and H. Kudo, Truncated Hilbert
transform and image reconstruction from limited tomographic data, Inverse Probl. 223, 10371053 2006.
53
Y. B. Ye, H. Y. Yu, Y. C. Wei, and G. Wang, A general local reconstruction approach based on a truncated Hilbert transform, Int. J. Biomed.
Imaging 2007, Article ID 63634, 8 pages 2007.
54
Y. B. Ye, H. Y. Yu, and G. Wang, Exact interior reconstruction with
cone-beam CT, Int. J. Biomed. Imaging 2007, Article ID 10693, 5 pages
2007.
55
F. Natterer, The Mathematics of Computerized Tomography Society for
Industrial and Applied Mathematics, Philadelphia, 2001.
56
G. Wang, Y. B. Ye, and H. Y. Yu, General VOI/ROI reconstruction
methods and systems using a truncated Hilbert transform, Patent disclosure submitted to Virginia Tech. Intellectual Properties on May 15 2007.
57
H. Y. Yu and G. Wang, A general formula for fan-beam lambda tomography, Int. J. Biomed. Imaging 2006, Article ID 10427, 9 pages 2006.
58
H. Y. Yu, Y. C. Wei, Y. B. Ye, and G. Wang, Lambda tomography with
discontinuous scanning trajectories, Phys. Med. Biol. 5214, 43314344
2007.
59
H. Y. Yu and G. Wang, A general scheme for velocity tomography,
Signal Process. 885, 11651175 2008.
60
D. Saintfelix et al., In-vivo evaluation of a new system for 3D computerized angiography, Phys. Med. Biol. 393, 583595 1994.
61
L. T. Niklason et al., Digital tomosynthesis in breast imaging, Radiology 2052, 399406 1997.
Medical Physics, Vol. 35, No. 3, March 2008
1063
62
W. Ross, D. D. Cody, and J. D. Hazle, Design and performance characteristics of a digital flat-panel computed tomography system, Med. Phys.
336, 18881901 2006.
63
S. Greschus et al., Potential applications off lat-panel volumetric CT in
morphologic and functional small animal imaging, Neoplasia 78, 730
740 2005.
64
F. Kiessling et al., Volumetric computed tomography VCT: A new
technology for noninvasive high-resolution monitoring of tumor angiogenesis, Nat. Med. 1010, 11331138 2004.
65
J. M. Boone et al., Computed tomography for imaging the breast, J.
Mammary Gland Biol. Neoplasia 112, 103111 2006.
66
B. Chen and R. Ning, Cone-beam volume CT breast imaging: Feasibility
study, Med. Phys. 295, 755770 2002.
67
L. T. Holly and K. T. Foley, Image guidance in spine surgery, Orthop.
Clin. North Am. 383, 451461 2007.
68
N. Clinthorne, Intra-operative imaging, J. Laparoendosc Adv. Surg.
Tech. A 155, 549555 2005.
69
W. Berninger and R. Redington, Multiple purpose high speed tomographic x-ray scanner, US Patent No. 4,196,352A 1980.
70
T. G. Flohr et al., First performance evaluation of a dual-source CT
DSCT system, Eur. Radiol. 162, 256268 2006.
71
Y. Kyriakou and W. A. Kalender, Intensity distribution and impact of
scatter for dual-source CT, Phys. Med. Biol. 5223, 69696989 2007.
72
J. Zhao, M. Jiang, T. Zhuang, and G. Wang, An exact reconstruction
algorithm for triple-source helical cone-beam CT, J. X-Ray Sci. Technol.
143, 191206 2006.
73
J. Zhao, M. Jiang, T. G. Zhuang, and G. Wang, Minimum detection
window and inter-helix PI-line with triple-source helical cone-beam scanning, J. X-Ray Sci. Technol. 142, 95107 2006.
74
J. Zhao, Y. N. Jin, Y. Lu, and G. Wang, A reconstruction algorithm for
triple-source helical cone-beam CT via filtered backprojection, 9th International Meeting on Fully Three-Dimensional Image Reconstruction in
Radiology and Nuclear Medicine, pp. 205208, Lindau, Germany 2007.
75
S. S. Orlov, Theory of three dimensional reconstruction I: Conditions for
a complete set of projections, Sov. Phys. Crystallogr. 20, 312314
1975.
76
B. De Man, P. Edic, and S. Basu, An iterative algorithm for timeresolved reconstruction of a CT scan of a beating heart, 8th Meeting on
Fully ThreeDimensional Image Reconstruction in Radiation and
Nuclear Medicine, Salt Lake City, UT, 356359 2005.
77
D. Boyd et al., High-speed, multi-slice, x-ray computed tomography,
Proceedings of SPIE, Vol. 372, pp. 139150, Pacific Grove, CA 1982.
78
G. Wang et al., Top-level design and preliminary physical analysis for
the first electron-beam micro-CT scanner, J. X-Ray Sci. Technol. 124,
251260 2004.
79
B. De Man et al., Stationary computed tomography system and method,
US Patent No. 7,280,631 B22005.
80
J. S. Price, W. Block, and M. Vermilyea, Extended multi-spot computed
tomography x-ray source, US Patent No. 6,983,035 B22006.
81
Z. Yin, B. De Man, and J. Pack, Analytical cone-beam reconstruction
using a multi-source inverse geometry CT system, Proceedings of SPIE,
Vol. 6510, San Diego, CA 2007.
82
T. G. Schmidt, R. Fahrig, N. J. Pelc, and E. G. Solomon, An inversegeometry volumetric CT system with a large-area scanned source: A feasibility study, Med. Phys. 319, 26232627 2004.
83
B. De Man et al., Multi-source inverse geometry CT: A new system
concept for X-ray computed tomography, Proceedings of SPIE, Vol.
6510, San Diego, CA 2007.
84
G. Wang and M. W. Vannier, Bolus-chasing angiography with adaptive
real-time computed tomography, U.S. Patent No. 6,535,821, allowed on
11/26/2002, issued on 3/18/2003 2003.
85
E. W. Bai et al., Study of an adaptive bolus chasing CT angiography, J.
X-Ray Sci. Technol. 141, 2738 2006.
86
A. Katsevich, S. Basu, and J. Hsieh, Exact filtered backprojection reconstruction for dynamic pitch helical cone beam computed tomography,
Phys. Med. Biol. 4914, 30893103 2004.
87
E. Tkaczyk et al., Simulation of CT dose and contrast-to-noise as function of bowtie shape, Proceedings of SPIE, Vol. 5368, pp. 403410, San
Diego, CA 2004.
88
M. Kalra, N. Naz, S. Rizzo, and M. Blake, Computed tomography radiation dose optimization: Scanning protocols and clinical applications of
automatic exposure control, Curr. Probl Diagn. Radiol. 345, 171181
2005.
1064
89
1064
2007.
T. Beyer et al., A combined PET/CT scanner for clinical oncology, J.
Nucl. Med. 418, 13691379 2000.
111
D. A. Boas and A. M. Dale, Simulation study of magnetic resonance
imaging-guided cortically constrained diffuse optical tomography of human brain function, Appl. Opt. 4410, 19571968 2005.
112
K. Kagawa et al., Initial clinical assessment of CT-MRI image fusion
software in localization of the prostate for 3D conformal radiation
therapy, Int. J. Radiat. Oncol., Biol., Phys. 382, 319325 1997.
113
G. Wang, Y. Li, and M. Jiang, Uniqueness theorems in bioluminescence
tomography, Med. Phys. 318, 22892299 2004.
114
J. Nuyts et al., Simultaneous maximum a posteriori reconstruction of
attenuation and activity distributions from emission sinograms, IEEE
Trans. Med. Imaging 185, 393403 1999.
115
A. Momose and J. Fukuda, Phase-contrast radiographs of nonstained rat
cerebellar specimen, Med. Phys. 224, 375379 1995.
116
R. A. Lewis, Medical phase contrast X-ray imaging: Current status and
future prospects, Phys. Med. Biol. 4916, 35733583 2004.
117
A. Momose, Recent advances in x-ray phase imaging, Jpn. J. Appl.
Phys., Part 1 449A, 63556367 2005.
118
X. Z. Wu and H. Liu, Clarification of aspects in in-line phase-sensitive
X-ray imaging, Med. Phys. 342, 737743 2007.
119
X. Z. Wu, H. Liu, and A. M. Yan, x-ray phase-attenuation duality and
phase retrieval, Opt. Lett. 304, 379381 2005.
120
S. W. Wilkins et al., Phase-contrast imaging using polychromatic hard
X-rays, Nature London 384, 335338 1996.
121
F. Pfeiffer, T. Weitkamp, O. Bunk, and C. David, Phase retrieval and
differential phase-contrast imaging with low-brilliance x-ray sources,
Nat. Phys. 24, 258261 2006.
122
C. Kottler, C. David, F. Pfeiffer, and O. Bunk, A two-directional approach for grating based differential phase contrast imaging using hard x
rays, Opt. Express 153, 11751181 2007.
123
M. Jiang, C. L. Wyatt, and G. Wang, X-ray phase-contrast imaging with
three 2D gratings, Int. J. Biomed. Imaging 2007, Article ID 27152
2007.
124
D. W. Holdsworth, Micro-CT in small animal and specimen imaging,
Trends Biotechnol. 208, S34S39 2002.
125
M. J. Paulus, A review of high-resolution x-ray computed tomography
and other imaging modalities for small animal research, Lab Anim. 30,
3645 2001.
126
G. Wang, Micro-CT scanners for biomedical applications: An overview, Adv. Imaging 16, 1827 2001.
127
G. Wang et al., Design, analysis and simulation for development of the
first clinical micro-CT scanner, Acad. Radiol. 124, 511525 2005.
128
D. J. Brenner, C. D. Elliston, E. J. Hall, and W. E. Berdon, Estimated
risks of radiation-induced fatal cancer from pediatric CT, AJR, Am. J.
Roentgenol. 1762, 289296 2001.
129
A. Berrington de Gonzalez and S. Darby, Risk of cancer from diagnostic
x rays: Estimates for the UK and 14 other countries, Lancet 363, 345
351 2004.
130
O. W. Linto and F. A. Mattler, Jr. , National conference on dose reduction in computed tomography, emphasis on pediatrics, AJR, Am. J.
Roentgenol. 181, 321329 2003.
131
G. Wang, S. Y. Zhao, and D. Heuscher, A knowledge-based cone-beam
x-ray CT algorithm for dynamic volumetric cardiac imaging, Med. Phys.
298, 18071822 2002.
110
Arjun G. Yodh
Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, Pennsylvania 191046396
and NCI Network for Translational Research in Optical Imaging, Beckman Laser Institute,
Irvine, California 92612
David A. Boas
NMR Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Boston, Massachusetts 02129 and NCI Network for Translational Research in Optical Imaging,
Beckman Laser Institute, Irvine, California 92612
Albert E. Cerussi
Beckman Laser Institute and Medical Clinic, University of California Irvine, Irvine, California 92612
and NCI Network for Translational Research in Optical Imaging, Beckman Laser Institute,
Irvine, California 92612
Received 25 January 2008; revised 2 April 2008; accepted for publication 3 April 2008;
published 22 May 2008
Diffuse optical imaging DOI is a noninvasive optical technique that employs near-infrared NIR
light to quantitatively characterize the optical properties of thick tissues. Although NIR methods
were first applied to breast transillumination also called diaphanography nearly 80 years ago,
quantitative DOI methods employing time- or frequency-domain photon migration technologies
have only recently been used for breast imaging i.e., since the mid-1990s. In this review, the state
of the art in DOI for breast cancer is outlined and a multi-institutional Network for Translational
Research in Optical Imaging NTROI is described, which has been formed by the National Cancer
Institute to advance diffuse optical spectroscopy and imaging DOSI for the purpose of improving
breast cancer detection and clinical management. DOSI employs broadband technology both in
near-infrared spectral and temporal signal domains in order to separate absorption from scattering
and quantify uptake of multiple molecular probes based on absorption or fluorescence contrast.
Additional dimensionality in the data is provided by integrating and co-registering the functional
information of DOSI with x-ray mammography and magnetic resonance imaging MRI, which
provide structural information or vascular flow information, respectively. Factors affecting DOSI
performance, such as intrinsic and extrinsic contrast mechanisms, quantitation of biochemical components, image formation/visualization, and multimodality co-registration are under investigation in
the ongoing research NTROI sites. One of the goals is to develop standardized DOSI platforms that
can be used as stand-alone devices or in conjunction with MRI, mammography, or ultrasound. This
broad-based, multidisciplinary effort is expected to provide new insight regarding the origins of
breast disease and practical approaches for addressing several key challenges in breast cancer,
including: Detecting disease in mammographically dense tissue, distinguishing between malignant
and benign lesions, and understanding the impact of neoadjuvant chemotherapies. 2008 American Association of Physicists in Medicine. DOI: 10.1118/1.2919078
I. INTRODUCTION
The field of optical imaging in medicine has seen continuous
growth for decades, and there is a wide array of technologies
and applications in development and clinical use. The range
of different optical hardware choices available leads to a
complicated process of convergence upon the optimal system
2443
for each specific clinical need. For example, even within the
categories of light sources, detectors, and light delivery, there
are many dozens of options to choose from, and the choices
made can drastically alter the data that is possible to be obtained. Two central benefits from optical imaging are the
ability to gain molecular-level information and submicro-
0094-2405/2008/356/2443/9/$23.00
2443
2444
Tromberg et al.: Diffuse optical imaging technologies for breast cancer management
Population
benefiting
Size of
Target cost
Clinical trial per scan
Screening
All
society
10,000s
Screening
high risk groups
Select
groups
1000s
Diagnosis following
abnormal mammogram
Integration into
clinical imaging systems
Monitoring
Therapy
Those
previously
screened
Cancer
patients
$100
500+
$1000
100s
$2,000+
scopic structural information, about the tissue function. Fundamental studies indicate that the molecular and cellular
specificity are possible, in both endogenous tissue markers
and with exogenous contrast agents. However, rapid medical
translation can be slowed by the lack of convergence to a
single technology, and yet strategic industry and academic
collaborations can be used to help advance translation. The
National Cancer Institute NCI has funded specific Networks for Translational Research in Optical Imaging
NTROI through a competitive review process, whose mission has been to define technologies which have the highest
potential promise for impact in cancer, and find ways to test
and validate them in multicenter trials. The Network preparing this report was funded to advance optical imaging technologies that have high potential to translate into successful
multicenter trials in a setting related to breast cancer management. The focus of this paper is to outline these promising technological innovations and look towards what development time scales they should be expected on.
In any imaging technology development, there is a key set
of trade-offs which are dictated largely by the application
needs. In Fig. 1, an illustration of the key needs in breast
cancer detection, diagnosis, and management are listed on
the left, along with estimates of the subject population who
would benefit from the system, and an order of magnitude
estimate of the scope of a clinical trial which would be
needed to demonstrate the system utility. Additionally, the
application area of the system will ultimately have a cost
which makes the system financially viable for the application. For example, population wide screening programs
would have widespread societal impact, yet would require
clinical trials with potentially tens of thousands of people to
prove the system efficacy, and ultimately could only be cost
effective if the cost per person was as low as mammography,
Medical Physics, Vol. 35, No. 6, June 2008
2444
2445
Tromberg et al.: Diffuse optical imaging technologies for breast cancer management
2445
FIG. 2. The Laser Breast Scanner LBS system developed at the University of California Irvine, Beckman Laser Institute, which has been produced for
multicenter trials of monitoring neoadjuvant chemotherapy response in breast cancer. The system electronics and console are shown in A, and the tissue
probe with light source fibers and light detectors in B, and the procedure to take measurements across the tumor region right breast and the controlateral
breast left breast are shown, with the probe being moved in 1 cm increments, to allow measurement of the on-tumor and off-tumor values of the tissue optical
index TOI.
2446
Tromberg et al.: Diffuse optical imaging technologies for breast cancer management
2446
2447
Tromberg et al.: Diffuse optical imaging technologies for breast cancer management
2447
FIG. 3. Tomographic imaging systems developed for breast tumor characterization and imaging during therapy have been developed and large clinical trial
results have been published. The commercial system from ART Inc. see Ref. 15 is shown in a photograph A, and research at Dartmouth resulted in the
system shown in B Refs. 14, 28, and 29, and a schematic of the system at the University of Pennsylvania is shown in C Refs. 6 and 30. Each system
uses multiple sets of measurements at multiple wavelengths of NIR light, transmitted through the breast to reconstruct images of hemoglobin, oxygen
saturation, water, and scattering values.
this imaging can be done readily with a visit taking less than
30 min. The utility of these systems in a truly widespread
screening application is still unproven, but clinical trials are
ongoing to assess sensitivity and specificity in subgroups.
IV. OPTICAL AS AN ADJUNCT IMAGING
TOOL
Optical imaging as an adjunct device to mammography or
ultrasound imaging is a natural choice, because the information gained with the optical signal is distinctly different from
that of the clinical imaging information. Near-infrared scanning of suspected abnormalities could provide the added information required to better determine the malignancy status,
Medical Physics, Vol. 35, No. 6, June 2008
and either help determine whether biopsy is needed, or ultimately track suspected abnormalities over time to detect malignant types of changes. There have been several commercial ventures aimed at this particular technology space in the
health care market, and two devices have gained approvals in
some countries and in an evaluation phase in the U.S., including ComfortScan ART Inc., Montreal Canada and
Computed Tomography Laser Mammography system
CTLM Imaging Diagnostic Systems, Inc., Plantation FL.
Research in this area has also been carried out by a European
consortium and by Philips Medical Systems. The key to success in this area has been to find an optimal way to maximize
the clinical use of information of the optical image alongside
2448
Tromberg et al.: Diffuse optical imaging technologies for breast cancer management
2448
(A)
(B)
(C)
FIG. 4. Hybrid Modality systems have been developed and used in pilot clinical trials to characterize tumors and test the synergy in having mutual information
and the ability to combine information data sets to create a new class of hybrid images. The University of Pennsylvania system was the first such device A
Refs. 26 and 27, and similar systems were built into a 3Tesla MRI at Dartmouth B Refs. 18 and 20, and into an x-ray tomosynthesis system at the
Massachusetts General Hospital C Ref. 31.
2449
Tromberg et al.: Diffuse optical imaging technologies for breast cancer management
NIH sponsored 3T MR-coupled system being tested currently B, and the NIR system built into an x-ray breast
tomosynthesis system C. While the primary screening modality for the breast is x-ray mammography, there has been
comparatively little study of how to introduce optics into this
modality, indicating that integration with the other modalities
would lead to applications which are not in the screening
area. Thus the potential for adding optics into a secondary
imaging modality then lies in the area of providing information which can differentiate the diagnosis of the lesion, or
perhaps track therapy response.
If the key role for optics integrated into clinical systems is
to provide functional information for a targeted region within
the breast, then the system integration needs to be carried out
to maximally utilize the information from the two systems in
a complementary or ideally synergistic manner. A key
method of integration is then to maximize the spectral content of the optical hardware and utilize the spatial prior relations from the clinical imaging system.1620 This approach to
integrating spectral constraints and spatial prior information
as a constraint has been demonstrated in tumor imaging,20
and can now be implemented with optimized software algorithms becoming available through the network. As illustrated in Fig. 1, DOS imaging within other imaging devices
would have a cost that was likely matched to the system it
was coupled to, such that DOS coupled to ultrasound would
likely have to be quite inexpensive to make an impact,
whereas DOS coupled MR could be considerably more expensive since the latter cost is limited by the MR mainly. The
economic feasibility of these hybrid systems is a little uncertain at present, however performance will likely have a major
influence on the cost which is appropriate.
Optical tomography has been integrated into a breast
x-ray tomosynthesis system Fig. 4C at the Massachusetts
General Hospital, and is undergoing clinical trials.21,22 Because of the widespread use of x-ray imaging of the breast,
this combination makes fundamental sense, and may provide
additional information where mammography has limited efficacy, such as in the dense breast or for distributed or complex lesions.
Additionally, soft tissue imaging with US is the natural
follow on procedure for certain diagnoses, and integration of
optical spectroscopy into this has been productive and led the
way towards region-based estimation of tissue
properties.2325 This approach makes sense for US, as the
region to be characterized is usually known when US is being used, and the goal is then to provide as much information
about the tumor as is feasible. The ability to image hemoglobin, oxygen saturation, and scatter has all been shown, with
strong potential for added diagnostic information.
The integration of diffuse tomography into MRI was a
key development which was initially pioneered at the University of Pennsylvania26,27 and this work is being carried on
to explore ways to synergize the mutual information through
the use of MR-derived spatial constraints in the diffuse optical imaging DOI spectroscopy,18,20 and the exponential
growth of breast MRI in recent years makes these developments of heightened importance Figs. 4A and 4B. With
Medical Physics, Vol. 35, No. 6, June 2008
2449
MR, there is increased ability to constrain the optical property recovery both spatially as well as in chromophore concentrations, as the MR can yield water and fat maps which
can be used to constrain the spectral recovery of hemoglobin,
oxygen saturation, scatter, and exogenous agents.
The integration of NIR into clinical imaging is a natural
step in the progression of integration of clinical information
into a streamlined series of information sources. One illustration of this with widespread acceptance is the newly used
hybrid PET/CT scanners, where the PET information has
gained much wider appreciation through the ability to superimpose the scan on the high resolution CT image, and further
to allow enhancement of the image quality through postprocessing based upon the CT. Similar to this, overlay of NIR
spectral data on the MR scans and enhancement of the NIR
spectral data quantitatively should allow easier clinical acceptance of this new information stream.
VI. CONCLUSIONS
The four applications listed here are key areas where the
healthcare need overlaps very well with the emerging technology which is available in the next decade. Each area requires refinement of the NIR technology or completion of
multicenter clinical trials to verify that the technology will
truly be a useful solution to the problem. NIR spectral tomography of tissue has had a rich history of development in
NIH funded work, and some of the designs discussed in this
overview are a result of decades of development. Advanced
stage clinical designs that will be advanced towards multicenter trials are desperately needed to carry out these validation trials. Coordination of these trials with the established
medical clinical trials bodies such as the National Cancer
Institutes Imaging Clinical Trials program or through the
American College of Radiology Imaging Network ACRIN
would be the ideal way to ensure that these technologies are
advanced appropriately with maximal overlap with other information sources.
In some cases the business model involved in the system
development can lead to success or failure of the technology,
however designs which have been vetted through both peerreviewed development as well as business plan evaluation
would be ideal to ensure that the right technology is advanced with the greatest promise for success. Ensuring that
academic-industry partnerships have a pathway for this cooperation is critical, in a manner which is not too encumbered by limited funding issues, but also provides room for
competition and intellectual property preservation. This is a
complex landscape in which to develop advanced technologies, but the National Cancer Institute has made the first
solid step in this process through sponsorship of the Network
for Translational Research in Optical Imaging. Development
of optical contrast agents which will provide true molecular
or compartmental specificity for cancer still remain to be
developed to the point of FDA approval and commercial
distribution, however large amounts of research are still under way in this area. It is likely that these exogenous contrast
agents will be used in breast cancer imaging or therapy, but
2450
Tromberg et al.: Diffuse optical imaging technologies for breast cancer management
the timeline for FDA approval for human use is still quite
uncertain at this time. The technologies outlined here for
intrinsic contrast imaging are capable and ready to be used in
molecular contrast imaging as well, so it is likely that after
the first round of multicenter trials begin in endogenous imaging, that this may facilitate the means for studying contrast
agents in a multicenter setting as well.
This paper is a summary report from one of these sponsored networks for translational research in optical imaging,
where the outcome of the network has been exactly focused
on providing a convergence in the space of advanced NIR
technology, breast cancer health care need, and business development capacity. The clinical trials proposed here will be
carried out over the next decade or so, and will likely have a
direct impact on both the technologies, businesses, and
medical practice involved in breast cancer detection and
management.
ACKNOWLEDGMENT
This work has been funded by the Network for Translational Research in Optical Imaging, within the National Cancer Institute U54CA105480.
a
2450
2451
Tromberg et al.: Diffuse optical imaging technologies for breast cancer management
U. L. Osterberg, and K. D. Paulsen, Quantitative hemoglobin tomography with diffuse near-infrared spectroscopy: Pilot results in the breast,
Radiology 2181, 261266 2001.
30
J. P. Culver, R. Choe, M. J. Holboke, L. Zubkov, T. Durduran, A. Slemp,
V. Ntziachristos, B. Chance, and A. G. Yodh, Three-dimensional diffuse
optical tomography in the parallel plane transmission geometry: Evalua-
2451
Received 6 December 2007; revised 8 April 2008; accepted for publication 8 April 2008;
published 27 May 2008
This is a review of methods, currently and potentially, available for significantly reducing x-ray
exposure in medical x-ray imaging. It is stimulated by the radiation exposure implications of the
growing use of helical scanning, multislice, x-ray computed tomography for screening, such as for
coronary artery atherosclerosis and cancer of the colon and lungs. Screening requires highthroughput imaging with high spatial and contrast resolution to meet the need for high sensitivity
and specificity of detection and classification of specific imaged features. To achieve this goal
beyond what is currently available with x-ray imaging methods requires increased x-ray exposure,
which increases the risk of tissue damage and ultimately cancer development. These consequences
limit the utility of current x-ray imaging in screening of at-risk subjects who have not yet developed
the clinical symptoms of disease. Current methods for reducing x-ray exposure in x-ray imaging,
mostly achieved by increasing sensitivity and specificity of the x-ray detection process, may still
have potential for an up-to-tenfold decrease. This could be sufficient for doubling the spatial
resolution of x-ray CT while maintaining the current x-ray exposure levels. However, a spatial
resolution four times what is currently available might be needed to adequately meet the needs for
screening. Consequently, for the proposed need to increase spatial resolution, an additional order of
magnitude of reduction of x-ray exposure would be needed just to keep the radiation exposure at
current levels. This is conceivably achievable if refraction, rather than the currently used attenuation, of x rays is used to generate the images. Existing methods that have potential for imaging the
consequences of refracted x ray in a clinical setting are 1 by imaging the edge enhancement that
occurs at the interfaces between adjacent tissues of different refractive indices, or 2 by imaging
the changes in interference patterns resulting from moving grids which alter the refraction of x rays,
that have passed through the body, in a predictable fashion, and 3 theoretically, by an image
generated from the change in time-of-flight of x-ray photons passing through the body. Imaging
phase shift or change in time-of-flight, rather than attenuation, of x-ray photons through tissues
presents formidable technological problems for whole-body 3D imaging. However, if achievable in
a routine clinical setting, these approaches have the potential for greatly expanding the use of x-ray
imaging for screening. This overview examines the increased contrast resolution and reduced radiation exposure that might be achievable by the above-mentioned methods. 2008 American
Association of Physicists in Medicine. DOI: 10.1118/1.2919112
Key words: screening, x-ray refractive index, x-ray attenuation
I. INTRODUCTION
Shortly after its introduction to clinical practice in 1973,
computed tomography was rapidly shown to greatly increase
the specificity and sensitivity of image information content.1
Since the mid 1990s fast, multislice, helical scanning multidetector, or multislice, computed tomography MDCT or
MSCT,2,3 which generates a 3D array of tiny cubic picture
elements voxels, has advanced to include screening applications. Examples include CT colonography,4 lung imaging
for early detection of cancer,5 and coronary artery
calcification.6
Because of the clinical appeal of these developments,
there is considerable pressure for further technological improvement to increase the spectrum of clinical applications,
especially in screening for early stages of disease. This, however, is likely to increase the radiation exposure involved,
2502
0094-2405/2008/356/2502/11/$23.00
2502
2503
ral relationship between radiation exposure and consequences. For instance, Imaizumi et al.7 recently showed that
a significant linear radiation dose response for thyroid nodules, including malignant tumors and benign tumors, exists
in the survivors of atom bomb exposure.
With the introduction of 3D imaging CT scanners, radiation exposures are increasingly being expressed as volume
computed tomography dose index CTDIvol in mGy and
dose-length product DLP in mGycm. Although rarely experienced in clinical diagnostic or interventional practices,
after a 2 Gy single dose, skin reddening occurs.8 However,
because not all tissues are equally radiosensitive, gonads, for
instance, are more sensitive than bone marrow colon
lung stomach and these are more sensitive than bladder,
liver, and thyroid, which in turn are more sensitive than skin.
A reasonable approximation of the more biologically meaningful effective dose E can be obtained by the equation
1
E = K DLP,
2
0.1 0.21020
0.21020 0.2220.1
0.2120.1 0.21020
20 0.2120.1
Ioe0.210
= 1.5 10 Io ,
5
2503
FIG. 1. K is the kinetic energy released in matter and is the area exposed.
Extrapolation of these data to 511 keV indicates that use of the positron
annihilation method would involve almost ten times higher KERMA than
that at 60 keV. With permission from Ref. 51.
Because the radiation exposure required to achieve a desired image quality depends so strongly on the desired image
resolution, the pathophysiological rationale for the level of
image resolution likely to be needed for screening purposes
needs to be established.
Screening for a disease requires both low false negative
and false positive detections as well as high true positive and
high true negative detections, i.e., high specificity and sensitivity. For imaging this requires spatial, contrast, and/or temporal resolutions depending on the type of pathological pro-
2504
2504
2505
2505
2506
2506
group = c2/phase ,
FIG. 2. Complex refractive index n of breast tissue, where is contribution by photon refraction and is contribution from attenuating factors.
Note that at 60 keV contributes 104 times the value of . Figure modified
with permission from Ref. 51.
ii
2507
A third, conceivable, approach is to cyclically modulate the illuminating x-ray flux with a cycle duration
i.e., wavelength very much longer e.g., 1010 than
an x-ray photon wavelength. The Moir pattern resulting from the refraction of these waves would be more
manageable in terms of fewer phase wraparounds.
The downside of this approach is that a sufficient
number of photons N would be needed, per modulation wavelength, to provide the needed sensitivity to
detect the small phase shift . To measure the
phase difference between the two trains of n x-ray
pulses for the purposes of illustration, each with a
Gaussian distribution of N photons, the number of
photons needed to detect a phase difference between the two identical trains of pulses is
= FWHM/2.355N n,
2507
2508
2508
FIG. 3. Two approaches to time-of-flight x-ray imaging. Left panel: a very brief x-ray pulse is launched and the change in time of arrival of the x ray at the
detector used to generate the image. Right panel: a positron annihilation results in two gamma ray photons which are launched simultaneously in opposite
directions. The difference in time of detection between the reference and imaging detector pair is the signal used to generate the image.
to have several hits before others have even one hit. If the
number of detected hits in a detection pixel follows a Poisson distribution with a mean dependent on the number of
photons that survive and are detected, then
Px = eNdNdx/x ! ,
FIG. 4. If each pixel in this 9-pixel detector array has an equal chance of
being hit by a random x-ray photon emitted from a point x-ray source, and
detection efficiency is 100%, then approximately double the number of photons are needed to be reasonably sure that each pixel receives at least one
photon. The panels show the distribution of the number of detected photons
that might occur after the array is exposed by the indicated number of
photons.
2509
2509
TABLE I. Each detector pixel should detect at least one photon to prevent dead spots in the image. The random spatial distribution of photons see Fig. 4,
attenuation of photon flux by the body, and the inefficiency of the detector combine to decrease the chance of a detector pixel detecting an x-ray photon. This
decrease is mitigated by increasing the x-ray flux destined for each typical detector pixel. The numbers in bold indicate those conditions for which less than
0.01% of detector pixels each detect less than five photons.
X-ray flux mean number of photons aimed
at 1 detector pixel
Efficiency of detector fraction of
transmitted photons detected by detector
Attenuation by body thickness
5 cm
10 cm
15 cm
20 cm
25 cm
60
80
0.5
0.7
0.9
0.95
0.5
0.7
0.09
0.95
0.4
9.7
41
73
97
0.01
1.4
18
52
87
0.00
0.14
6.4
32
62
0.00
0.00
1.53
19
52
0.00
0.00
0.18
8
36
0.00
0.00
0.10
6
32
0.5
0.7
100
0.9
0.95
10
2510
2510
FIG. 6. If the x-ray detection process of a detector is cut off after a short
time interval from a selected time zero, then the total number of photons
area under curve detected decreases with delay of the x-ray pulse left
panel. Hence, the difference in the area under the curves is related both to
delay of the pulse due to any difference in attenuation right panel within
the subject.
2511
2511
TABLE II. For the positron annihilation approach a pair of photons is emitted in opposite directions from a
source of finite thickness such that the arrival times of the photons at the detector pixel has a Gaussian
distribution. As the number of detected event pairs is increased, the discrimination of time differences t
between the reference detector pixel and imaging detector pixel can be a fraction of the time resolution t
of the clock used to measure the times of the event occurrences.
# Observed events pairs
Discrimination t / t
2
0.75
4
0.55
or its potential for resolution down to 100 m without increasing the radiation exposure beyond that currently experienced at 600 m voxel resolution.
ACKNOWLEDGMENTS
The author thanks Professor Dr. Ulrich Bonse, Technical
University Dortmund, Dortmund, Germany; Professor Dr.
Winfried Schuelke, Institute of Physics, University of Dortmund, Dortmund, Germany; Professor James L. Ritman,
University of Bochum and Research Center Jlich, Institute
for Nuclear Physics, Jlich, Germany, for their input on some
of the x-ray/matter interaction issues; Dr. Vernon S. Pankratz
and Mr. Jeffrey Slezak of the Mayo Clinic Biostatistics Section for performing the statistical analysis underlying the
analysis illustrated in Tables I and II, and Ms. Mara Lukenda
for typing and formatting this manuscript.
a
8
0.4
16
0.3
32
0.2
64
0.15
128
0.10
2512
2512
13611368 2004.
G. Woan, The Cambridge Handbook of Physics Formulas Cambridge
University Press, Cambridge, 2000, p. 219.
48
Van Nostrands Scientific Encyclopedia, 5th ed., edited by D. M. Considine Van Nostrand Reinhold Co., New York, 1976.
49
A. Momose and J. Fukuda, Phase-contrast radiographs of nonstained rat
cerebellar specimen, Med. Phys. 22, 375379 1995.
50
T. J. Davis, D. Gao, T. E. Gureyev, A. W. Stevenson, and S. W. Wilkins,
Phase-contrast imaging of weakly absorbing materials using hard
x-rays, Nature London 373, 595598 1995.
51
R. A. Lewis, Medical phase contrast x-ray imaging: Current status and
future prospects, Phys. Med. Biol. 49, 35733583 2004.
52
T. Weitkamp, C. David, C. Kottler, O. Bunk, and F. Pfeiffer, Tomography with grating interferometers at low-brilliance sources, Proc. SPIE
6318, 63180S 2006.
53
T. Takeda, A. Momose, J. Wu, Q. Yu, T. Zeniya, T.-T. Lwin, A.
Yoneyama, and Y. Itai, Vessel imaging by interferometric phase-contrast
x-ray technique, Circulation 105, 17081712 2002.
54
F. Beckmann, U. Bonse, F. Busch, and O. Gnnewig, X-ray microtomography microCT using phase contrast for the investigation of organic
matter, J. Comput. Assist. Tomogr. 21, 539553 1997.
55
T. Weitkamp, A. Diaz, B. Nhammer, F. Pfeiffer, T. Rohbeck, P. Cloetens,
M. Stampanoni, and C. David, Hard x-ray phase imaging and tomography with a grating interferometer, Proc. SPIE 5535, 137142 2004.
56
G. E. Moore, Cramming more components onto integrated circuits,
Electronics 38, 114117 1965.
57
C. Martel, S. Fourmaux, L. Lecherbourg, H. Bandulet, and J. C. Kieffer,
New ultra fast x-ray streak camera for the advanced laser light source
facility, Proc. SPIE 6703, 67030K 2007.
58
H. Shimizu, Y. Iikura, Y. Sasano, and N. Takeuchi, Resolution improvement in an analog-to-digital converter by the superposed dither signal,
Electron. Commun. Jpn. 64, 18 1981.
47
Gerald L. DeNardob
Internal Medicine, University of California Davis Medical Center, 1508 Alhambra Boulevard, Suite 3100,
Sacramento, California 95816
Ruby F. Meredithc
Department of Radiation Oncology, Wallace Tumor Institute WTI No. 117, University of Alabama
at Birmingham, Birmingham, Alabama 35294
Received 4 March 2008; revised 22 April 2008; accepted for publication 10 May 2008;
published 12 June 2008
Targeted radionuclide therapy TRT seeks molecular and functional targets within patient tumor
sites. A number of agents have been constructed and labeled with beta, alpha, and Auger emitters.
Radionuclide carriers spanning a broad range of sizes; e.g., antibodies, liposomes, and constructs
such as nanoparticles have been used in these studies. Uptake, in percent-injected dose per gram of
malignant tissue, is used to evaluate the specificity of the targeting vehicle. Lymphoma B-cell has
been the primary clinical application. Extension to solid tumors will require raising the macroscopic
absorbed dose by several-fold over values found in present technology. Methods that may effect
such changes include multistep targeting, simultaneous chemotherapy, and external sequestration of
the agent. Toxicity has primarily involved red marrow so that marrow replacement can also be used
to enhance future TRT treatments. Correlation of toxicities and treatment efficiency has been limited by relatively poor absorbed dose estimates partly because of using standard phantom organ
sizes. These associations will be improved in the future by obtaining patient-specific organ size and
activity data with hybrid SPECT/CT and PET/CT scanners. 2008 American Association of
Physicists in Medicine. DOI: 10.1118/1.2938520
Key words: radionuclide therapy, antibodies, absorbed dose
I. PRESENT CLINICAL SITUATION
A possible imaging result with a cancer patient is shown in
Fig. 1, whereby multiple lesions appear using nuclear or
other technology. It is important to find a method to target
both seen and unseen tumor locations anywhere in the body.
It would be desirable that this strategy be as specific as possible to the tumor with minimal collateral toxicity. Targeting
of this sort is measured in units of percent-injected dose per
gram of tissue % ID/ g. The mouse would generally be the
test species for preclinical development of radiopharmaceuticals for targeted radionuclide therapy TRT. Following IV
injection into a 20 g animal, nonspecific uptake should be
100% ID/20 g = 5% ID/g
0094-2405/2008/357/3062/7/$23.00
3062
3063
3063
D = S A ,
FIG. 1. Anterior gamma camera image of a medullary thyroid cancer patient. Image obtained at 48 h post-IV injection of 5 mCi 185 MBq of
111
In-cT84.66 anti-CEA antibody. Multiple bone lesions are seen in the pelvis and both femurs. Part of the right lobe of liver appears superiorly see
Ref. 37.
the liver. Intravenous injections have been limited to treatment of lymphoma using anti-B cell anti-CD20 monoclonal
antibodies.
II. CURRENT LIMITATIONS
Because of the small number of FDA-approved agents, it
is clear that significant constraints have hindered development of TRT. Primarily, the limitation has been relatively
poor tumor uptake. This has led to correspondingly low absorbed doses and lack of tumor response. Table III contains
estimated absorbed doses for a standard phase III lymphoma
trial14 and an experimental colon cancer therapy.16 These
protocols were at previously determined maximal values of
activity per body mass14 or per body surface area.16 Notice
that both lymphoma and colon cancer regimens have
achieved similar numerical tumor absorbed dose values.
Clinical application of TRT to the former disease state has
followed from the relatively high sensitivity of B-cells to
ionizing radiation.
NaI
MIBGa
Octreotideb
SHALsc
Nucleotidesd
Antibodiese
Liposomesf
Nanoparticlesg
Morpholinosh
Spheresi
154 Da
130 Da
100 Da
2 kDa
10 kDa
25150 kDa
100 nm
10 nm
2 kDa
30 m
Application
Representative radiolabels
131
See Ref. 3.
See Ref. 4.
c
See Ref. 5.
d
See Ref. 6.
e
See Ref. 7.
Medical Physics, Vol. 35, No. 7, July 2008
See Ref. 8.
See Ref. 9.
h
See Ref. 10.
i
See Refs. 11 and 12.
I
I
90
Y, 111In
131
I
99m
Tc, 111In
90
Y and 131I
111
In, 99mTc
131
188
Re
Y
90
3064
3064
Label
Disease
Method
90
90
where f is a fraction on the order of 0.3 and the ratio represents the correction for marrow mass 1500 g in a particular
standard phantom20 in a patient with an assumed 5000 g
total blood mass. In the ideal case of no direct marrow targeting of the radioactive agent or its label, the result of Eq.
3 is used as an input into Eq. 2 to estimate total RM
absorbed dose.
Issues of accurate absorbed dose estimation have been at
the heart of TRT since its inception in the 1950s; e.g., there is
little knowledge of the absorbed dose to remnant thyroid
tissue during 131I treatment. While MIRD-type calculations
are often used to justify clinical trials to the FDA, use of the
associated phantom mass values contained in programs like
MIRDOSE3 Ref. 20 and OLINDA Ref. 21 cannot be directly
applied to a given patient. It has been shown, for example,
that hepatic and splenic masses may differ by factors of twoand threefold in individuals undergoing TRT of colon
cancer.22 Patient-specific treatment planning23 requires actual
knowledge of patient anatomy organ separations and organ
sizes masses. This problem is thus one of image fusion
whereby the activity found using quantitative nuclear imaging can be assigned correctly to a particular anatomic
structure.
III. FUTURE DEVELOPMENTS
Medical physicists can be expected to contribute directly
to the growth of TRT in the next 710 years. Advances
would be of two distinctly different but complementary
types. Initially there will have to be a strategy for develop-
ing, testing, and generally enhancing the tumor-targeting capability of radiopharmaceuticals. Computer modeling of biodistributions and comparisons of figures of merit are aspects
of this work relevant to the physicist. This is particularly
important for solid tumors. Beyond the engineering and inventive phase will be the necessity of establishing improved
methods of patient-specific absorbed dose calculations.
These estimates will become more useful when they relate to
voxels rather than whole organ values as indicated above.
Among target tissue calculations, red marrow absorbed dose
estimates will continue to be the most important.
III.A. Pharmaceutical development and testing
TABLE III. Comparison of two TRT clinical antibody protocols involving 90Y. Absorbed doses were computed using the MIRD formalism Refs. 20 and 21
and refer to whole organ mean values. Ranges are shown in parentheses. The clinical protocols were at maximum activity per body mass Ref. 14 or body
surface area Ref. 16 determined from previously established hematological toxicity levels.
Agent
Zevalina
Ant-CEAb
Disease
NHL
Colon cancer
RM absorbed dose
cGy
1484
61 24 300
1320
46 6400
71
18 221
64
19 198
532
234 1856
912
534 1719
3065
3065
ciently depleted. It has become conventional to replace platelets and white cells in patients undergoing TRT when these
cells are below certain limits. In the future, such procedures
may become economical enough such that the marrow is no
longer the activity-limiting tissue. In that case, other solid
organs such as the kidneys or liver may be considered limiting. This strategy may lead to greater total activities being
injected and larger tumor absorbed doses being achieved.
As in the case of external beam, one anticipates that considerable effort will be expended to enhance TRT-induced
absorbed dose by means of circulating chemotherapeutic and
radiation-enhancing agents.28 At present, it is unclear what
chemical dose levels and what timing relative to the IV injection of the radiotherapeutic agent are optimal. Again, preclinical and phase I trials will be needed to evaluate the
effectiveness of this approach. Because of the multiple possible combinations of enhancing and TRT agents, extensive
animal testing will become more important in the near term.
Following from the above multiple agent therapies, it is
probable that future collaborations would be expanded between medical oncology and radiation oncology. Historically,
chemotherapeutic drugs have been studied using only blood
and excreta chemical measurements. In the next 710 years,
it would be anticipated that present-day and future chemotherapeutic drugs would be radiolabeled and then followed,
probably with PET scanning, in patients. Because of concern
that the radioactive tag may affect the biodistribution, labeling will preferably be done by substituting radioactive isotopes into the chemical structure. While organic isotopes 11C,
15
O, and 13N are of primary importance, there will be other
possible labels such as 18F in the case of 5FU. Availability of
local cyclotrons is implicit in this approach due to the short
half-lives of the positron labels required.
III.B. Improved absorbed dose estimation
3066
3066
IV. CONCLUSIONS
Continued growth in lymphoma treatment using TRT is
expected with more patients being entered into RIT trials as
part of a first-line therapy. Helping to expedite this expansion
is the unexplained, growing incidence of B-cell lymphomas.
We would anticipate that T-cell antibodies would become
available within the next 5 years. While 90Y facilitates outpatient treatments, the utilization of 131I as a label is expected to continue due to lower radionuclide cost and the
possibility of directly imaging therapeutic administrations to
verify treatment planning.
With better dosimetry, repeat TRT procedures would become possible. In the next 57 years, repeated lymphoma
therapies would become a topic of importance to patients
who have recurrence or were originally undertreated. Patientspecific normal organ absorbed doses would be tabulated to
make sure that the relevant total is within the levels determined in external beam therapy.17 It is likely that the toxicity
levels due to TRT-derived absorbed doses will be found to be
different from those seen in external beam therapies.
We can also expect extensive use of engineering and invention to contribute a number of new targeting
pharmaceuticals.36 Improvement in chelates will enable
greater control of radiometals so that direct targeting of the
freed label to the marrow becomes improbable.
Improved patient-specific absorbed dose estimates will
lead to better control of toxicities and improved clinical results for lymphomas and, eventually, solid tumors. The
present tenuous connection between estimated marrow absorbed dose and its clinical toxicity will become more statistically significant as the marrow mass becomes available via
marrow tracers used as adjuncts prior to the TRT protocols.
As a result, escalation in phase I and other FDA-approved
trials would occur with normal organ absorbed doses as the
control variables and not simplistic parameters such as total
activity or total activity per body surface area or body mass.
Labeling and tracking of chemotherapeutics will evolve out
of this work and lead to greater understanding and safety of
their administration to individual patients. Combinations of
radiation-enhancing compounds with TRT will expand
within these two converging threads of knowledge. Patientspecific treatment is one logical outcome of this work.
ACKNOWLEDGMENTS
This work was partially supported by NIH Grant Nos.
PO1-43904 and CA 33572 L.W..
a
3067
3067
J. Macey, and K. R. Lamborn, Factors affecting 131-I-Lym-1 pharmacokinetics and radiation dosimetry in patients with non-Hodgkins lymphoma and chronic lymphocytic leukemia, J. Nucl. Med. 40, 13171326
1999.
19
J. A. Siegel, B. W. Wessels, E. E. Watson, M. G. Stabin, H. M. Vriesendorp, E. W. Bradley, C. C. Badger, A. B. Brill, C. S. Kwok, D. R. Stickney, K. F. Eckermann, D. R. Fisher, D. J. Buchsbaum, and S. E. Order,
Bone marrow dosimetry and toxicity in radioimmunotherapy, Antibody, Immunoconjugates, Radiopharm. 3, 213233 1990.
20
M. G. Stabin, MIRDOSE: Personal computer software for internal absorbed dose assessment in nuclear medicine, J. Nucl. Med. 37, 538546
1996.
21
M. G. Stabin, R. B. Sparks, and E. Crowe, OLINDA/EXM: The secondgeneration personal computer software for internal absorbed dose assessment in nuclear medicine, J. Nucl. Med. 46, 10231027 2005.
22
L. E. Williams, A. Liu, A. A. Raubitschek, and J. Y. C. Wong, A method
for patient-specific absorbed dose estimation for internal beta emitters,
Clin. Cancer Res. 5, 3015s3019s 1999.
23
G. Akabani, W. G. Hawkins, M. B. Eckblade, and P. K. Leichner,
Patient-specific dosimetry using quantitative SPECT imaging and threedimensional discrete Fourier transform convolution, J. Nucl. Med. 38,
308314 1997.
24
C. H. Holdsworth, M. Dahlbom, A. Liu, L. Williams, C. S. Levin, M.
Janecek, and E. J. Hoffman, Expanding the versatility of a more accurate
accelerated Monte Carlo simulation for 3D PET: Data correction of PET
emission scans using 124-I, Nuclear Science Symposium Conference
Record 2001 IEEE 2002, Vol. 4, pp. 21052109.
25
D. A. Goodwin, Tumor pretargeting: Almost the bottom line, J. Nucl.
Med. 36, 876879 1995.
26
D. M. Goldenberg and R. M. Sharkey, Novel radiolabeled antibody conjugates, Oncogene 26, 37343744 2007.
27
O. Linden, J. Kurkus, M. Garkavij, E. Cavallin-Stahl, M. Ljungberg, R.
Nilsson, T. Ohlsson, B. Sandberg, S.-E. Strand, and J. Tennvall, A novel
platform for radioimmunotherapy: Extracorporeal depletion of biotinylated and 90Y-labeled rituximab in patients with refractory B-cell lymphoma, Cancer Biother. Radiopharm. 20, 457466 2005.
28
S. Welt, G. Ritter, C. Williams, Jr., L. S. Cohen, A. Jungbluth, E. A.
Richards, L. J. Old, and N. E. Kemeny, Preliminary report of a phase I
study of combination chemotherapy and humanized A33 antibody immunotherapy in patients with advanced colorectal cancer, Clin. Cancer Res.
9, 13471353 2003.
29
L. E. Williams, A. Liu, D. M. Yamauchi, G. Lopatin, A. A. Raubitschek,
and J. Y. C. Wong, The two types of correction of absorbed dose estimates for internal emitters, Cancer 94, 12311234 2002.
30
D. J. Macey, L. E. Williams, H. B. Breitz, A. Liu, T. K. Johnson, and P.
B. Zanzonico, A Primer for Radioimmunotherapy and Radionuclide
Therapy AAPM Report No. 71 Medical Physics, Madison, WI, 2001.
31
K. F. Koral, Y. Dewaraja, L. A. Clarke, J. Li, R. Zasadny, S. G. Rommelfanger, I. R. Francis, M. S. Kaminski, and R. L. Wahl, Tumor-absorbed
dose estimates versus response in tositumomab therapy of previously untreated patients with follicular non-Hodgkins Lymphoma: Preliminary
report, Cancer Biother. Radiopharm. 15, 347355 2000.
32
C. Hindorf, O. Linden, L. Stenberg, J. Tennvall, and S.-E. Strand,
Change in tumor-absorbed dose due to decrease in mass during fractionated radioimmunotherapy in lymphoma patients, Clin. Cancer Res. 1,
4003S4006S 2003.
33
L. E. Williams, R. B. Duda, R. T. Proffitt, B. G. Beatty, J. D. Beatty, J. Y.
C. Wong, J. E. Shively, and R. J. Paxton, Tumor uptake as a function of
tumor mass: A mathematic model, J. Nucl. Med. 29, 103109 1988.
34
D. J. Macey, S. J. DeNardo, G. DeNardo, J. L. Goodnight, and M. W.
Unger, Uptake of In-111 labeled monoclonal antibody ZME018 as a
function of tumor size in a patient with melanoma, Am. J. Physiol.
Imaging 3, 16 1988.
35
G. Riethmuller, E. Schneider-Gadicke, G. Schlimok, W. Schmiegel, R.
Raab, K. Hoffken, R. Gruber, H. Pichlmaier, H. Hirche, R. Pichlmayr, P.
Buggisch, and J. Witte, Randomised trial of monoclonal antibody for
adjuvant therapy of resected DukesC colorectal carcinoma, Lancet 343,
11771183 1994.
36
J. Carlsson, E. Aronsson Forsell, S. O. Hietala, T. Stigbrand, and T.
3068
3068
Received 14 April 2008; revised 7 May 2008; accepted for publication 8 May 2008;
published 16 June 2008
Photodynamic therapy PDT is an emerging treatment modality that employs the photochemical
interaction of three components: light, photosensitizer, and oxygen. Tremendous progress has been
made in the last 2 decades in new technical development of all components as well as understanding of the biophysical mechanism of PDT. The authors will review the current state of art in PDT
research, with an emphasis in PDT physics. They foresee a merge of current separate areas of
research in light production and delivery, PDT dosimetry, multimodality imaging, new photosensitizer development, and PDT biology into interdisciplinary combination of two to three areas. Ultimately, they strongly believe that all these categories of research will be linked to develop an
integrated model for real-time dosimetry and treatment planning based on biological
response. 2008 American Association of Physicists in Medicine. DOI: 10.1118/1.2937440
Key words: PDT, spectroscopy, implicit dosimetry, explicit dosimetry, dynamic process
I. INTRODUCTION
Photodynamic therapy PDT is an emerging cancer treatment modality based on the interaction of light, a photosensitizing drug, and oxygen.1 PDT has been approved by the
U.S. Food and Drug Administration for the treatment of microinvasive lung cancer, obstructing lung cancer, and obstructing esophageal cancer, as well as for premalignant actinic keratosis and age-related macular degeneration. Studies
have shown some efficacy in the treatment of a variety of
malignant and premalignant conditions including head and
neck cancer,2,3 lung cancer,46 mesothelioma,7 Barretts
esophagus,8,9 prostate,1012 and brain tumors.9,1315 Unlike radiation therapy, PDT uses nonionizing radiation and can be
administered repeatedly without cumulative long-term complications since it does not appear to target DNA.
There has been tremendous progress made in the last 2
decades in new technologies and in understanding of the
basic biophysical mechanisms of PDT. The most important
question to be answered is: What determines PDT efficacy
for a particular patient, photosensitizer, and treatment protocol? Answering this question will require a unified understanding of the interactions of the three basic components:
light, photosensitizer, and tissue oxygenation. We have categorized the current basic research in PDT into five areas: 1
light sources, light transport, and light delivery in tissue; 2
PDT dosimetry; 3 optical and anatomic imaging; 4 new
photosensitizers; and 5 PDT biology. Among these, the development of new photosensitizers and PDT biology is traditionally considered outside of the realm of PDT physics
and will only be briefly described for completeness. All five
areas are linked by a quantitative understanding of the dynamic processes involved in the photochemical interaction
that drives PDT. In the next section, we will describe these
areas separately.
3127
0094-2405/2008/357/3127/10/$23.00
3127
3128
ISC
S1
3128
T1
1
Photon
absorption
S0
O2
Sensitizer
O2
Oxygen
FIG. 1. Energy level diagram for a typical type II photosensitizer and oxygen. The sensitizer in its ground state S0 absorbs a photon of light and is
excited to its first singlet state S1. It spontaneously decays to its excited
triplet state T1 via ISC. From T1, energy is transferred to ground state
molecular oxygen 3O2, creating reactive singlet oxygen 1O2.
3129
sensitive chromophores. Implicit dosimetry31 uses a quantity such as fluorescence photobleaching of the photosensitizer, which is indirectly predictive of the production of
singlet oxygen. Strategies for direct and implicit dosimetry
are under development and will be discussed in later sections.
II.B.3. Anatomic and optical imaging
The most commonly used medical imaging modalities include ultrasound, computer tomography CT, magnetic
resonance, magnetic resonance spectroscopy, single photon
emission computer tomography, and positron emission tomography PET. The first three modalities produce excellent
anatomical images, while the latter three provide functional
information e.g., oxygen perfusion or tissue metabolism at
the expense of image resolution. Diffuse optical tomography
DOT is a viable new biomedical imaging modality.36 This
technique images the absorption and scattering properties of
biological tissues and has been explored as a diagnosis tool
in breast,3742 brain,43,44 and bones and joints.45 Some preliminary attempts have been made to perform DOT for
prostate.4649 DOT at the treatment wavelength can be used
as input to calculate light fluence rate distribution for PDT.
In addition, it provides access to a variety of physiological
parameters that cannot otherwise be measured easily. Another modality, optical coherence tomography OCT uses
coherent light to obtain high resolution images. However,
OCT is rarely used in PDT because of the limitation of penetration depth 1 mm.50
Most image reconstruction of DOT is based on solving
the inverse problem for the diffusion equation.27 This is an
ill-posed problem because of the strong scattering in the turbid medium. As a result, the image resolution of DOT is
limited when compared with other imaging modalities, such
as magnetic resonance imaging MRI or CT. The use of
spectroscopic information and/or a priori anatomic information is common to provide additional constraint to produce
reliable DOT reconstruction. Interested readers can find more
information from some excellent review articles.51,52
The concept of absorption and fluorescence spectroscopy
as a modality for the diagnosis of disease dates back several
decades.53 In recent years, the potential for spectroscopy to
diagnose cancer54 and to monitor the progress of cancer
treatment55 has been increasingly appreciated. In addition to
its role in diagnosis, spectroscopy is particularly applicable
to PDT in determining the local drug and oxygen
concentrations.56
Photodynamic therapy can cause changes in the concentration and oxygenation of blood in tissue both directly,
through photochemical oxygen consumption, and indirectly,
through effects on the vasculature and general physiological
responses. The monitoring of these responses may, therefore,
be predictive of treatment outcome. For oxygen monitoring,
the difference in absorption spectra between oxy- and deoxyhemoglobin is used to determine the hemoglobin oxygen
saturation, i.e., the fraction of total hemoglobin that is in its
oxygenated state. This quantity can be related to the oxygen
Medical Physics, Vol. 35, No. 7, July 2008
3129
concentration in the blood using the Hill curve.57 This measurement does not directly measure the concentration of oxygen in the tissue itself, however, it is possible to model the
relationship between the vascular and tissue oxygenation.
This concept has been investigated in animal models in
which the blood flow and/or oxygenation were monitored
and changes correlated with outcome.58,59
II.B.4. New photosensitizers
Various photosensitizing drugs have been developed. The
first-generation photosensitizer, hematoporphyrin derivative,
is a mixture of porphyrin monomers and oligomers that is
partially purified to produce the commercially available
product, porfimer sodium, marketed under the tradename
Photofrin. Photofrin was approved for treatment of early
stage lung cancer in 1998 and for Barretts esophagus in
2003. The clinical applicability of Photofrin has been limited by two factors. First, its absorption peak occurs at too
short a wavelength 630 nm to allow deep penetration in
tissue. Second, administration of Photofrin results in cutaneous photosensitivity lasting up to 6 weeks. These limitations have inspired the development of a second generation
of photosensitizers with longer-wavelength absorption peaks
and more rapid clearance from skin. Among these was benzoporphyrin derivative monoacid A BPD-MA, or verteporfin. In preclinical trials, it was observed that verteporfin preferentially targeted neovasculature. This selectivity has been
exploited for the treatment of choroidal neovascularization
CNV, an abnormal growth of vessels in the retina associated with age-related macular degeneration AMD, the leading cause of blindness in the developed world. Verteporfin
was approved in the U.S. under the tradename Visudyne for
CNV treatment in 2000. Tetra m-hydroxyphenyl chlorin
mTHPC, Foscan is another second generation photosensitizer, a pure synthetic chlorin compound, which is activated
by 652 nm light.60 The major advantages of mTHPC are a
short duration of skin photosensitivity 15 days, a high
quantum yield for singlet oxygen, and depth of tumor necrosis of up to 10 mm in preclinical models.61 mTHPC has been
used for treatment of pleural mesothelioma,7 head-and-neck
cancers,62,63 esophagus,64,65 prostate,10,66 pancreas,67 arthritic
joints,68 and skin cancers69 and was approved in Europe for
PDT of head-and-neck and varieties of other tumors in 2001.
Another development of note is the prodrug
-aminolevulinic acid ALA. Unlike other PDT drugs, ALA
itself is not a photosensitizer. When taken up by cells, however, it is converted by a naturally occurring biosynthetic
process into the photosensitizer protoporphyrin IX PpIX.
ALA can be applied topically, and was approved by the FDA
in 1999 for the treatment of actinic keratosis AK. Table I
summaries several of the more widely used photosensitizers
currently available.
II.B.5. PDT biology
From the point of view of biological response, PDT is
fundamentally different from other cancer therapies. Unlike
ionizing radiation, PDT achieves its cytotoxic effects prima-
3130
3130
Photosensitizer
Porfimer sodium
ALA-PpIX
Methyl aminolevulate-PpIX
Hexyl aminolevulate-PpIX
BPD-MA
mTHPC
Trade name
Approval
Excitation Drug-light
nm
interval
Clearance
time
Photofrin
Levulan Keratastick
Metvix
Hexvix
Verteporfin, Visudyne
Foscan
630
405, 635
405, 635
405
689
652
48150 h
1418 h
3h
13 h
15 min
48110 h
46 weeks
2 days
2 days
2 days
5 days
15 days
Phase I trials
Phase I trials
Phase I and II trials
732
762
664
3h
30 min
1h
2 h
Phase I trials
672
2436 h
Motexafin Lutetium
MLu, Lutex, Lutrin
Pd-bacteriopheophorbide
Tookad
Taloporfin sodium
LS11
mono-L-aspartyl chlorin e6
Silicon pthalocyanine 4
PC-4
Light Delivery
Dosimetry
Light Dosimetry
with Feedback
Complex
Dosimetry with
Feedback,
Real-time TPS
long times after injection.72 An additional level of complexity arises from the fact that the response to PDT is not confined to the cells where the singlet oxygen is deposited but
can involve physiological73 and immunological74,75 responses as well.
III. CURRENT DEVELOPMENTS
Much of the research in the early decades of PDT and its
related fields proceeded in five almost independent areas, as
illustrated in the top row of Fig. 2. The problems associated
with light source and light delivery system development, dosimetry, and optical imaging were treated as physics problems, while photosensitizer development and PDT biology
were treated as problems of chemistry and biology, respectively. In recent years, however, the most promising ad-
Photosensitizer
Development
Imaging
Explicit and,
Implicit
Dosimetry,
SOLD
Sites
Biomarkers
Molecular
Beacons
PMB, doseresponsive
imaging
PDT
Biology
Targeted
Photosensitizer,
Photoimmunotherapy
Biological
Response
Imaging
Real-time dosimetry
and treatment planning
based on biological
response
FIG. 2. Diagram illustrating the progress of PDT development from a set of disparate fields top row to a collaborative effort unifying the contributions of
biologists, chemists, physicists, and engineers. The second row illustrates the current state of art of research and represents integration of two separate fields.
The third row illustrates the future research direction and the fourth row is the ultimate integration of all disparate fields. See text for a complete description.
Medical Physics, Vol. 35, No. 7, July 2008
3131
3131
Because the goal of PDT is ultimately to deliver the optimal treatment to the tumor, the idea of using noninvasive
detection and imaging to determine the status of tumor cells
at the molecular level is very attractive. This concept has
driven the development of molecular beacons, molecules that
target specific molecular pathways that can be imaged using,
for instance, near-infrared fluorescence. Steflova et al. have
developed a series of molecular beacons whose fluorescence
3132
Several researchers have developed methods for enhancing the ability of a photosensitizer to target tumors. One
strategy to accomplish this is to optimize the timing the administration of light to coincide with the desired distribution
of the photosensitizer.72 This approach has been used to target vasculature by applying light while the photosensitizer is
in circulation, for the treatment of AMD using Vertepofin and
the prostate using Tookad.12 In cases where the rate of production of singlet oxygen is limited by the vascular resupply
of oxygen, changes in the light fluence rate can change the
distribution of deposited singlet oxygen, with higher fluence
rates leading to preferential targeting of vascular-adjacent tissue. Thus, the combination of drug-light interval and fluence
rate can be adjusted to enhance the treatment of the desired
tissue type.
A more direct approach to photosensitizer targeting is the
conjugation of the photosensitizer to a tumor-selective molecule or particle.91 The last 2 decades have seen tremendous
progress in the development of tumor-targeted therapeutic
and imaging agents for cancer in general.92,93 These innovations will continue to inform the development of new photosensitizers targeted to specific tumor types.
Recent research has shown PDT induced increases of
VEGF, MMPs, and/or COX-2 in tumor microenvironment
that cause resistance to standard treatment. As a result, combination of therapy that suppresses these growth factors has
been proposed to enhance the therapeutic efficacy of PDT.
C225, a monoclonal antibody that inhibits the receptor tyrosine kinase activity of EGFR, has been shown to enhance
the PDT treatment in ovarian cancer.94,95 Other combined
modalities seek to target treatment-induced angiogenesis
and/or inflammation to enhance the effectiveness of PDT.96
IV. FUTURE DEVELOPMENTS AND
PREDICTIONS
We predict that future developments in PDT will continue
the trend toward interdisciplinary work and the inclusion of
more technologies and subfields, such as imaging, novel
drug design, and biological modeling into the treatment planning and dosimetry processes. Below are a few predictions of
future PDT physics research directions:
IV.A. Light sources: Faster, cheaper, larger, smaller
3132
3133
under fluorescence imaging.103 Pandey et al.104 have proposed combining photosensitizers with contrast agents for
optical, MRI and PET imaging, allowing image-guided PDT
treatment. Zheng et al. have combined the molecular beacon concept with a photosensitizer to produce photodynamic molecular beacons.105 These molecules exhibit
quenching that suppresses their production of 1O2 in their
latent state. They can be activated by interaction with a
tumor-specific molecular marker, in this case the matrix metalloproteinase MMP-7. The clinical implementation of strategies such as these has the potential to dramatically improve
the targeting of the photosensitizer to the tumor. An essential
component of any targeting strategy will be the ability to
verify the targeting of the drug to the desired target. The
distribution of the photosensitizer can be determined optically using fluorescence or absorption imaging. In addition,
detailed models of the distributions and kinetics of light,
photosensitizer, and oxygen will provide a quantitative relationship between the microscopic distribution and photochemical properties of the drug and the macroscopically observed treatment response.106 In cases where the drug is
activated by an endogenous agent, the results will be predictable only using such sophisticated models.
IV.D. Treatment planning integrated with dosimetry,
imaging, and light delivery devices to allow
adaptive treatment
3133
tion of photosensitizer, it does not consider the effect of tissue oxygenation on the quantum yield of oxidative radicals.
Thus, it is only applicable in cases where ample oxygen supplies exist. It is anticipated that new dosimetry quantities
based on reacted singlet oxygen 1O2rx, e.g., a product of
singlet oxygen quantum yield and PDT dose, can be used to
account for kinetics of the oxygen consumption during PDT
process.23 Foster et al. have determined most of fundamental
parameters necessary for microscopic model of the oxygen
consumption during PDT.18,82,106,112 However, when applying models developed in the microscopic scale to a macroscopic environment, the values of many parameters may
change, and many parameters may be observable only in the
volume average, so extensive study will be required to determine the values for each specific photosensitizer.
IV.F. Physiological effects of PDT will be exploited to
develop systemic therapy
3134
3134
Photobiol. Sci. 4, 438442 2005.
M. J. Paterson, O. Christiansen, F. Jensen, and P. R. Ogilby, Overview of
theoretical and computational methods applied to the oxygen-organic
molecule photosystem, Photochem. Photobiol. 82, 11361160 2006.
17
T. H. Foster, D. F. Hartley, M. G. Nichols, and R. Hilf, Fluence rate
effects in photodynamic therapy of multicell tumor spheroids, Cancer
Res. 53, 12491254 1993.
18
I. Georgakoudi, M. G. Nichols, and T. H. Foster, The mechanism of
Photofrin photobleaching and its consequences for photodynamic dosimetry, Photochem. Photobiol. 65, 135144 1997.
19
S. L. Gibson, K. R. VanDerMeid, R. S. Murant, R. F. Raubertas, and R.
Hilf, Effects of various photoradiation regimens on the antitumor efficacy of photodynamic therapy for R3230AC mammary carcnimoas,
Cancer Res. 50, 72367241 1990.
20
B. W. Henderson, T. M. Busch, L. A. Vaughan, N. P. Frawley, D. Babich,
T. A. Sosa, J. D. Zollo, A. S. Dee, M. T. Cooper, D. A. Bellnier, W. R.
Greco, and A. R. Oseroff, Photofrin photodynamic therapy can significantly deplete or preserve oxygenation in human basal cell carcinomas
during treatment, depending on fluence rate, Cancer Res. 60, 525529
2000.
21
B. W. Henderson, T. M. Busch, and J. W. Snyder, Fluence rate as a
modulator of PDT mechanisms, Lasers Surg. Med. 38, 489493 2006.
22
X. H. Hu, Y. Feng, J. Q. Lu, R. R. Allison, R. E. Cuenca, G. H. Downie,
and C. H. Sibata, Modeling of a type II photofrin-mediated photodynamic therapy process in a heterogeneous tissue phantom, Photochem.
Photobiol. 81, 14601468 2005.
23
T. C. Zhu, J. C. Finlay, X. Zhou, and J. Li, Macroscopic modeling of the
singlet oxygen production during PDT, Proc. SPIE 6427, 642708
6427202007.
24
B. C. Wilson and M. S. Patterson, in Photodynamic Therapy of Neoplastic Disease, edited by D. Kessel CRC Press, Boca Raton, 1990, Vol. 1,
pp. 129144.
25
P. R. Almond, Photodynamic Therapy Equipment and Dosimetry American Institute of Physics, Woodbury, NY, 1992.
26
A. Ishimaru, Wave Propagation and Scattering in Random Media IEEE,
New York, 1997.
27
W. Cong, L. V. Wang, and G. Wang, Formulation of photon diffusion
from spherical bioluminescent sources in an infinite homogeneous medium, Biomed. Eng. Online 3, 12 2004.
28
R. C. Haskell, L. O. Svaasand, T.-T. Tsay, T.-C. Feng, M. S. McAdams,
and B. J. Tromberg, Boundary conditions for the diffusion equation in
radiative transfer, J. Opt. Soc. Am. A 11, 27272741 1994.
29
L. Wang, S. L. Jacques, and L. Zheng, MCML-Monte Carlo modeling of
light transport in multi-layered tissues, Comput. Methods Programs
Biomed. 47, 131146 1995.
30
J. J. Duderstadt and W. R. Martin, Transport Theory Wiley, New York,
1979.
31
B. C. Wilson, M. S. Patterson, and L. Lilge, Implicit and explicit dosimetry in photodynamic therapy: A new paradigm, Lasers Med. Sci. 12,
182199 1997.
32
S. K. Hendren, S. M. Hahn, F. R. Spitz, T. W. Bauer, S. C. Rubin, T. Zhu,
E. Glatstein, and D. L. Fraker, Phase II trial of debulking surgery and
photodynamic therapy for disseminated intraperitoneal tumors, Ann.
Surg. Oncol. 8, 6571 2001.
33
J. P. Marijnissen and W. M. Star, Performance of isotropic light dosimetry probes based on scattering bulbs in turbid media, Phys. Med. Biol.
47, 20492058 2002.
34
T. G. Vulcan, T. C. Zhu, C. E. Rodriguez, A. Hsi, D. L. Fraker, P. Baas,
L. H. Murrer, W. M. Star, E. Glatstein, A. G. Yodh, and S. M. Hahn,
Comparison between isotropic and nonisotropic dosimetry systems during intraperitoneal photodynamic therapy, Lasers Surg. Med. 26, 292
301 2000.
35
J. P. Marijnissen and W. M. Star, Calibration of isotropic light dosimetry
probes based on scattering bulbs in clear media, Phys. Med. Biol. 41,
11911208 1996.
36
A. H. Hielscher, A. Y. Bluestone, G. S. Abdoulaey, A. D. Klose, J.
Lasker, M. Stewart, U. Netz, and J. Beuthan, Near-infrared diffuse optical tomography, Dis. Markers 18, 313337 2002.
37
A. Li, E. L. Miller, M. E. Kilmer, T. J. Brukilacchio, T. Chaves, J. Stott,
Q. Zhang, T. Wu, M. Chorlton, R. H. Moore, D. B. Kopans, and D. A.
Boas, Tomographic optical breast imaging guided by three-dimensional
mammography, Appl. Opt. 42, 51815190 2003.
38
J. P. Culver, R. Choe, M. J. Holboke, L. Zubkov, T. Durduran, A. Slemp,
16
3135
3135
59
3136
80
3136
dosimetry system for prostate photodynamic therapy, Proc. SPIE 6845,
68450Q68458Q 2008.
99
L. Vesselov, W. Whittington, and L. Lilge, Design and performance of
thin cylindrical diffusers created in Ge-doped multimode optical fibers,
Appl. Opt. 44, 27542758 2005.
100
A. Rendon, R. Weersink, and L. Lilge, Towards conformal light delivery
using tailored cylindrical diffusers: attainable light dose distributions,
Phys. Med. Biol. 51, 59675975 2006.
101
G. Yu, T. Durduran, C. Zhou, T. C. Zhu, J. C. Finlay, T. M. Busch, S. B.
Malkowicz, S. M. Hahn, and A. G. Yodh, Real-time in situ monitoring
of human prostate photodynamic therapy with diffuse light, Photochem.
Photobiol. 82, 12791284 2006.
102
E. Clo, J. W. Snyder, P. R. Ogilby, and K. V. Gothelf, Control and
selectivity of photosensitized singlet oxygen production: challenges in
complex biological systems, ChemBioChem 8, 475481 2007.
103
Y. Chen, A. Gryshuk, S. Achilefu, T. Ohulchansky, W. Potter, T. Zhong,
J. Morgan, B. Chance, P. N. Prasad, B. W. Henderson, A. Oseroff, and R.
K. Pandey, A novel approach to a bifunctional photosensitizer for tumor
imaging and phototherapy, Bioconjug. Chem. 16, 12641274 2005.
104
R. K. Pandey, L. N. Goswami, Y. Chen, A. Gryshuk, J. R. Missert, A.
Oseroff, and T. J. Dougherty, Nature: A rich source for developing multifunctional agents. Tumor-imaging and photodynamic therapy, Lasers
Surg. Med. 38, 445467 2006.
105
G. Zheng, J. Chen, K. Stefflova, M. Jarvi, H. Li, and B. C. Wilson,
Photodynamic molecular beacon as an activatable photosensitizer based
on protease-controlled singlet oxygen quenching and activation, Proc.
Natl. Acad. Sci. U.S.A. 104, 89898994 2007.
106
S. Mitra and T. H. Foster, Photophysical parameters, photosensitizer
retention and tissue optical properties completely account for the higher
photodynamic efficacy of meso-tetra-hydroxyphenyl-chlorin vs Photofrin, Photochem. Photobiol. 81, 849859 2005.
107
A. Johansson, J. Axelsson, S. Andersson-Engels, and J. Swartling, Realtime light dosimetry software tools for interstitial photodynamic therapy
of the human prostate, Med. Phys. 34, 43094321 2007.
108
T. C. Zhu, J. Li, J. C. Finlay, A. Dimofte, D. Stripp, B. S. Malkowicz, and
S. M. Hahn, In vivo light dosimetry of interstitial PDT of human prostate, Proc. SPIE 6139, 61390L61401L 2006.
109
J. Li and T. C. Zhu, Determination of in vivo light fluence distribution in
heterogeneous prostate during photodyanmic therapy, Phys. Med. Biol.
53, 21032114 2008.
110
M. D. Altschuler, T. C. Zhu, J. Li, and S. M. Hahn, Optimized interstitial
PDT prostate treatment planning with the Cimmino feasibility algorithm,
Med. Phys. 32, 35243536 2005.
111
M. S. Patterson, B. C. Wilson, and R. Graff, In vivo tests of the concept
of photodynamic threshold dose in norma rate liver photosensitized by
aluminum chlorosulphonated phthalocyanine, Photochem. Photobiol. 51,
343349 1990.
112
I. Georgakoudi and T. H. Foster, Singlet oxygen- versus nonsinglet
oxygen-mediated mechanisms of sensitizer photobleaching and their effects on photodynamic dosimetry, Photochem. Photobiol. 67, 612625
1998.
113
A. P. Castano, P. Mroz, and M. R. Hamblin, Photodynamic therapy and
anti-tumour immunity, Nat. Rev. Cancer 6, 535545 2006.
114
S. O. Gollnick, L. Vaughan, and B. W. Henderson, Generation of effective antitumor vaccines using photodynamic therapy, Cancer Res. 62,
16041608 2002.
115
M. Korbelik and J. Sun, Photodynamic therapy-generated vaccine for
cancer therapy, Cancer Immunol. Immunother. 55, 900909 2006.
116
B. W. Henderson, S. O. Gollnick, J. W. Snyder, T. M. Busch, P. C. Kousis, R. T. Cheney, and J. Morgan, Choice of oxygen-conserving treatment regimen determines the inflammatory response and outcome of photodynamic therapy of tumors, Cancer Res. 64, 21202126 2004.
Received 18 February 2008; revised 29 May 2008; accepted for publication 29 May 2008;
published 8 July 2008
Molecular imaging MI constitutes a recently developed approach of imaging, where modalities
and agents have been reinvented and used in novel combinations in order to expose and measure
biologic processes occurring at molecular and cellular levels. It is an approach that bridges the gap
between modalities acquiring data from high e.g., computed tomography, magnetic resonance
imaging, and positron-emitting isotopes and low e.g., PCR, microarrays levels of a biological
organization. While data integration methodologies will lead to improved diagnostic and prognostic
performance, interdisciplinary collaboration, triggered by MI, will result in a better perception of
the underlying biological mechanisms. Toward the development of a unifying theory describing
these mechanisms, medical physicists can formulate new hypotheses, provide the physical constraints bounding them, and consequently design appropriate experiments. Their new scientific and
working environment calls for interventions in their syllabi to educate scientists with enhanced
capabilities for holistic views and synthesis. 2008 American Association of Physicists in Medicine. DOI: 10.1118/1.2948321
Key words: molecular imaging, data integration, unification
I. INTRODUCTION
Over the last 30 years we have experienced a rapid evolution
of technology which has led to a transformation of biological
and medical sciences. Biological entities and mechanisms
that the limitations of technology until recently did not allow
us to observe have now become an object of routine measurement and reasoning. A typical field of application of this
change is medical imaging. Until the early 1970s, the study
of the human body was based on imaging modalities that
produced projections, exploiting a variety of physical properties of the human tissues. Three dimensional 3D representations were rarely achieved through time-consuming and
mainly experimental procedures, producing rather vague final results. In contrast, modern routine medical diagnostic
imaging heavily relies on modalities that produce highresolution 3D representations of the human body featuring
both morphological and functional characteristics. The latter
often require four dimensional images, by combining the
three dimensional spatial information with other types of information, e.g., temporal, spectroscopic, etc.
Although computed tomography CT and magnetic resonance imaging MRI were initially considered to be inefficient, as it is vividly described in a recent article by Wagner,1
they proved to be indispensable medical diagnostic modalities. These modalities are mathematically ill-conditioned
problems. As such, much experimentation and cultivation of
ideas was necessary to make them appropriate for providing
clinically useful images. Instrumentation for functional tomographic imaging evolved along a path distinct from that of
anatomical imaging devices CT and MRI. The first human
tomographic images with positron-emitting isotopes PET
were presented in 1972,2 followed by single photon emission
3444
0094-2405/2008/358/3444/9/$23.00
3444
3445
Nikiforidis, Sakellaropoulos, and Kagadis: Molecular imaging and the unification of multilevel mechanisms
3445
TABLE I. The advent of MI allows the design of structured experiments aiming to reveal/find the causal relationships among variables and mechanisms
concerning various levels of biological organization. A unified biomedical theory may be built using top-down and bottom-up approaches as well as
combinations thereof for diagnosis or prognosis. This theory could causally connect the mutations of specific genes sequence analysis with a characteristic
pattern of gene coexpressions analysis of microarrays, the abnormal value of some immunochemical parameters flow cytometry, microscopy, the manifestation of certain macroscopic anatomical or functional anomalies CT, SPECT, etc. and finally with the observation of specific clinical signs and symptoms.
All modalities are in vivo unless otherwise indicated.
3446
Nikiforidis, Sakellaropoulos, and Kagadis: Molecular imaging and the unification of multilevel mechanisms
gregation of semantically similar data from multiple heterogeneous sources vertical integration; the composition of semantically complementary data from multiple heterogeneous
sources horizontal integration; and the standardization of
access to semantically similar information at disparate
sources integration for application portability. For example,
with vertical integration morphology information can be obtained through the combination of data from both CT and
ultrasound US; horizontal integration would be beneficial
when both anatomy and physiology of a specific region are
sought for. Integration for application portability would
make comparable data originating from imaging apparatuses
of different specifications, e.g., 512 512 and 256 256 image matrices.
The attempts for data integration have comprised a strenuous track of research within the field of medical informatics
and a number of issues still need to be addressed. Some of
these issues are the partial availability of databases, the controversial privacy issues, the fact that useful information lies
in the natural language text of the scientific papers, and, perhaps most importantly, the lack of appropriate standards.19 It
was soon evident that obtaining multiple representations of
the same entity, e.g., through CT, SPECT, or MRI, would
enhance the ability of making better medical decisions. But
the efficient combination of available variables toward better
medical diagnosis and prognosis had been a high-complexity
process, even when the available data were of the high-level
type, i.e., results of clinical and laboratory examinations.
With the advent of molecular data, the combination of
multimodality information reached a higher level of complexity. It became obvious that analytical approaches are unable to compensate for such complexity. From the thousands
of variables stored in the integrated databases only the variables relevant to specific clinical problems should be considered. This calls for the development of efficient ways for
variable description and management. One such way is
building additional data structurescalled metadatathat
describe the characteristics of stored variables. Metadata arranged in schemata can represent the hierarchical structures
of these characteristics and help us build frameworks for the
integration of contextual information from the clinicians
point of view. Statistical modeling and procedures for statistical learning can also be helpful, providing methodologies
for feature extraction and data reduction.
Evidence-based medicine EBM has been recently established as the approach of choice to medical practice. EBM
uses the best current evidence and statistical methods to justify the value of therapeutic decisions in an individual patient. What it effectively does is to identify similarities or
matches between the specific patients patterns of clinical
and laboratory data and scientifically valid evidence. The
availability of integrated data will allow us to describe all the
relevant variables and their interactions in a deep and structured manner and finally to expose the underlying mechanisms that give rise to these patterns. We could then obtain
better specifications of ones health status and consequently
aim at personalized treatment or prevention.
Medical Physics, Vol. 35, No. 8, August 2008
3446
3447
Nikiforidis, Sakellaropoulos, and Kagadis: Molecular imaging and the unification of multilevel mechanisms
3447
Instruments based on THz spectrometers with resolution better than 400 m are expected to complement existing biomedical imaging technologies.
Nanoparticles have the potential to change the role of imaging technologies in diagnosis and therapy. They are colloidal vesicular systems that vary in size from 10 to 1000 nm
with the drug and/or imaging probe of interest either entrapped therein or attached thereon.31 Inherently, when
echogenic nanoparticles are bound to surfaces, they can be
modified for compatibility with MR, US, x ray, or nuclear
imaging methodologies.32 There are several examples that
prove the advantages of such bimodal agents. For example,
fluorine 19F MRI of cells labeled with different types of
liquid perfluorocarbon nanoparticles produces unique and
sensitive cell markers distinct from any tissue background
signal.33 Successful full body microSPECT/CT mouse imaging of Cd125mTe/ ZnS NPs linked to either a monoclonal antibody against mouse lung thrombomodulin mAb 201B, or
a control antibody mAb 33, has shown that nanoparticles
conjugated to mAb 201B principally target the lungs while
the nanoparticles coupled to mAb 33 accumulate in the liver
and spleen.34 Similarly, PET labeled nanoparticles have been
successfully imaged.35 PET and MRI studies are carried out
in parallel due to the lack of combined imaging systems.36,37
It is estimated that simultaneous imaging of bimodal nanoparticles will maximize the benefits from the combination of
nuclear medicine and MR information.38 Other magnetic
nanoparticles can be caused to oscillate under the influence
of an incident ultrasonic wave with the advantage of greater
sensitivity due to the absence of a large background signal.39
By the use of targeted gold nanoparticles, the combination of
US with contrast enhanced photoacoustic imaging seems
possible. This is proposed as a visual tool to compound molecular and structural information for early stage prostate
cancer detection.40
An additional promise of endogenous expression, where
contrast reagents are engineered to interrogate endogenously
expressed proteins or nucleic acids, is the combination of
imaging with therapeutic reagents such that they can both be
optimized in an iterative fashion. The effect of therapeutic
reagents can give rise to modified signals which in turn can
be identified with the use of different contrast reagents. The
latter can give evidence for new therapeutic pathways, etc. It
is now recognized that many of the survival attributes of
neoplastic cells are determined by proteins involved in
mechanisms of antiapoptosis, cell cycle regulation, and damage repair.41 Therapeutic radiationfollowing the definition
of the underlying molecular features of cancer cells by MI
can act as focused biology i.e., producing molecular events
in the irradiated tissue that targets molecular entities other
than DNA, offering new opportunities for therapeutic attacks
on cancer cells.42
IV. UNIFICATION
In its course through history research often passes through
stages of conformity. In our opinion, we are currently experiencing such a stage. New research hypotheses are rareon
3448
Nikiforidis, Sakellaropoulos, and Kagadis: Molecular imaging and the unification of multilevel mechanisms
3448
3449
Nikiforidis, Sakellaropoulos, and Kagadis: Molecular imaging and the unification of multilevel mechanisms
3449
FIG. 1. The case of RA where various imaging modalities are used to provide details at different levels of information.
3450
Nikiforidis, Sakellaropoulos, and Kagadis: Molecular imaging and the unification of multilevel mechanisms
3450
modality covers efficiently only a narrow region. As technology advances, each modality becomes increasingly better in
terms of precision within this continuum, shedding light to
new mechanisms and processes. We are thus given the opportunity to build new theories that link the mechanisms relevant to one level with mechanisms relevant to another.
The design of new complex experiments will be needed,
involving variables pertinent to many levels of biological
organization. Obviously, MI is an interdisciplinary approach
where end-user needs and challenges have to be well defined
and clearly understood; clinicians define the diagnostic or
therapeutic needs; biologists can define the biological phenomena that can be targeted; and physicists will have to investigate the appropriate physical processes and suggest imaging concepts. These will lead to the advancement of both
science and technology. New theories will be developed and
new conceptual frameworks will be established to include
them. We thus have the opportunity to build genuine interdisciplinary collaborations within which the ideas originating
from different disciplines will fuse and create a new core of
thinking.
This is expected to affect the field of medical physics in
its theoretical foundations. It is unlikely that its current structure can accommodate the increasing interactions with biology and medicine; new ways of thinking will be needed to
meet new scientific challenges. We now have the possibility
of performing a step into revolutionary science,78 exploring
alternatives to long-held, obvious-seeming assumptions.
From an epistemological viewpoint, there are many similarities with the situation in physics at the dawn of the 20th
century. Experimental data that could not be explained with
the physical theories available at that time were partially accommodated by the Bohr atomic model and later with refinements made by Sommerfeld. However, only when the concept of the wave function and the Schrdinger equation
appeared did quantum theory manage to describe and explain
the mechanisms giving rise to the phenomena. The growth of
the quantum theory offered a unifying framework for concepts initially bound to either the microscopic or macroscopic level and gave birth to electronics as a new discipline.
Medical physics might follow the same route. Its theoretical
expansion may develop as a result of our attempt to provide
a unifying view of the biological processes from the molecule to the entire organism. MI may become both a technology that produces experimental data not yet accounted for by
current theories and a bridge through which fragments of
knowledge will be assembled into a unified theory.
The role that medical physicists will play in this endeavor
cannot be predicted precisely. It will be the result of a number of factors ranging from the reorientation of medical
physics courses syllabi to the self-motivation and degree of
engagement of each individual medical physicist. Medical
physicists should closely collaborate with biologists, biochemists, physicians, and bioinformaticists. They should take
a strategic position due to their diverse role in both providing
new results in the imaging research as well as giving feedback to the research of other disciplines.
3451
Nikiforidis, Sakellaropoulos, and Kagadis: Molecular imaging and the unification of multilevel mechanisms
3451
3452
Nikiforidis, Sakellaropoulos, and Kagadis: Molecular imaging and the unification of multilevel mechanisms
46
3452
315318 2008.
J. K. Jackson, T. Higo, W. L. Hunter, and H. M. Burt, Topoisomerase
inhibitors as anti-arthritic agents, Inflamm. Res. 57, 126134 2008.
62
H. Okamoto, Y. Katagiri, A. Kiire, S. Momohara, and N. Kamatani, Serum amyloid A activates nuclear factor-kappaB in rheumatoid synovial
fibroblasts through binding to receptor of advanced glycation endproducts, J. Rheumatol. 35, 752756 2008.
63
U. J. Netz, J. Beuthan, and A. H. Hielscher, Multipixel system for gigahertz frequency-domain optical imaging of finger joints, Rev. Sci. Instrum. 79, 034301 2008.
64
X. Wang, D. L. Chamberland, P. L. Carson, J. B. Fowlkes, R. O. Bude, D.
A. Jamadar, and B. J. Roessler, Imaging of joints with laser-based photoacoustic tomography: an animal study, Med. Phys. 33, 26912697
2006.
65
R. G. Cowell, A. P. Dawid, and S. L. Lauritzen, Probabilistic Networks
and Expert Systems Springer, Berlin, 1999.
66
F. V. Jensen and T. D. Nielsen, Bayesian Networks and Decision Graphs,
2nd ed. Springer, New York, 2007.
67
R. E. Neapolitan, Learning Bayesian Networks Pearson Prentice-Hall,
Englewood Cliffs, NJ, 2004.
68
J. Pearl, Probabilistic Reasoning in Intelligent Systems Morgan Kaufmann, San Francisco, CA, 1988.
69
D. Poole, Logic, knowledge representation, and Bayesian decision
theory, in Computational LogicCL 2000 Springer, Berlin/Heidelberg,
2000, Vol. 1861/2000, pp. 7086.
70
N. Cruz-Ramirez, H. G. Acosta-Mesa, H. Carrillo-Calvet, L. A. NavaFernandez, and R. E. Barrientos-Martinez, Diagnosis of breast cancer
using Bayesian networks: A case study, Comput. Biol. Med. 37, 1553
1564 2007.
71
J. P. Sacha, L. S. Goodenday, and K. J. Cios, Bayesian learning for
cardiac SPECT image interpretation, Artif. Intell. Med. 26, 109143
2002.
72
G. C. Sakellaropoulos and G. C. Nikiforidis, Development of a Bayesian
network for the prognosis of head injuries using graphical model selection
techniques, Methods Inf. Med. 38, 3742 1999.
73
Bayesian Networks for Expression References, http://www.stat.wisc.edu/
~yandell/statgen/reference/network.html, accessed January 31, 2008.
74
R. Jansen, H. Yu, D. Greenbaum, Y. Kluger, N. J. Krogan, S. Chung, A.
Emili, M. Snyder, J. F. Greenblatt, and M. Gerstein, A Bayesian networks approach for predicting protein-protein interactions from genomic
data, Science 302, 449453 2003.
75
S. M. McMahon, Quantifying the community: Using Bayesian learning
networks to find structure and conduct inference in invasions biology,
Biol. Invasions 7, 833844 2005.
76
C. J. Needham, J. R. Bradford, A. J. Bulpitt, and D. R. Westhead, A
primer on learning in Bayesian networks for computational biology,
PLOS Comput. Biol. 3, e129 2007.
77
S.Y. Kim, in Computational Methods in Systems Biology Springer, Berlin, 2003, Vol. 2602, pp. 104113.
78
T. S. Kuhn, The Structure of Scientific Revolutions The University of
Chicago Press, Chicago, 1962.
61
Received 2 March 2008; revised 10 July 2008; accepted for publication 10 July 2008;
published 16 September 2008
Nanostructures represent a promising new type of contrast agent for clinical medical imaging
modalities, including magnetic resonance imaging, x-ray computed tomography, ultrasound, and
nuclear imaging. Currently, most nanostructures are simple, single-purpose imaging agents based
on spherical constructs e.g., liposomes, micelles, nanoemulsions, macromolecules, dendrimers, and
solid nanoparticle structures. In the next decade, new clinical imaging nanostructures will be
designed as multi-functional constructs, to both amplify imaging signals at disease sites and deliver
localized therapy. Proposals for nanostructures to fulfill these new functions will be outlined. New
functional nanostructures are expected to develop in five main directions: Modular nanostructures
with additive functionality; cooperative nanostructures with synergistic functionality; nanostructures activated by their in vivo environment; nanostructures activated by sources outside the patient;
and novel, nonspherical nanostructures and components. The development and clinical translation
of next-generation nanostructures will be facilitated by a combination of improved clarity of the in
vivo imaging and biological challenges and the requirements to successfully overcome them; development of standardized characterization and validation systems tailored for the preclinical assessment of nanostructure agents; and development of streamlined commercialization strategies and
pipelines tailored for nanostructure-based agents for their efficient translation to the clinic. 2008
American Association of Physicists in Medicine. DOI: 10.1118/1.2966595
Key words: medical imaging, nanostructures, nanotechnology, contrast agents
I. INTRODUCTION
Imaging is a fundamental tool in the practice of modern
medicine. In parallel with the development of more sensitive
detectors and imaging systems, there is increasing interest in
designing new types of contrast agents for all modalities, to
sensitively and successfully diagnose specific disease pathologies. Exogenous contrast agents are designed to differentially scatter, absorb, or emit radiation such that upon introduction into a patient, the contrast agents location may be
distinguished from its surrounding tissue or background
noise. This may greatly increase the utility of imaging, as
normal and diseased tissues typically exhibit minimal native
contrast. Since the in vivo distribution of the contrast agent
depends on the patients physiology and the properties of the
agent, the radiologist can use contrast agents to reveal structural, functional, and/or molecular information about the patient to assist in disease diagnosis. At the forefront of new
contrast agent development are nanometer-scale structures,
or nanostructures, that can act as, or carry, contrast agents.
Nanostructures are constructs with physical dimensions
between a few nanometers to hundreds of nanometers in size.
Although simple nanostructures have been utilized for decades in clinical medicine e.g., colloidal and particulate carriers for pharmaceutical applications,1,2 and radiocolloids for
imaging and therapy3, there has been a recent revival in
developing new nanostructures of different materials, sizes,
and properties for biotechnological and medical applications
and, in particular, for medical imaging. Much of this re4474
0094-2405/2008/3510/4474/14/$23.00
4474
4475
4475
4476
Another key feature of medical nanostructures is their potential to carry significant payloads of small molecular or
particulate imaging agents within them or on their surface.
For example, a nanostructure 100 nm in diameter has a
volume equivalent to 100 000 small imaging molecules
and a surface area equivalent to the cross-sectional area of
10 000 small molecules. The localization of high concentrations of imaging agents within or on the surface of nanostructures combined with their preferential accumulation in
target sites can enhance the image signal-to-noise ratio, reduce dosage, and limit toxicity by decreasing distribution of
the imaging nanostructure in normal tissue.28
In addition to the EPR effect, nanostructures can potentially be designed to actively segregate to diseased tissue
or indicators of disease, through their conjugation to targeting moieties e.g., peptides, aptamers, or antibodies that specifically bind to surface epitopes or receptors preferentially
overexpressed at disease sites.2932 This type of active targeting may enhance the accumulation of nanostructures to disease sites, which is the reason behind the proposed use of
nanostructures for future molecular imaging applications.
Nanostructures may also be used to revive small molecule
agents that were previously limited by their innate nonideal
in vivo properties. For example, the incorporation of imaging
agents within nanostructures may increase the stability of
existing small molecule agents with short half-lives and
rapid clearance12,14,16,28 and allow molecular agents previously limited by their poor aqueous solubility to be used in
imaging and therapy applications.12,16,28,3335
II.B. Rational design of nanoscale contrast agents
4476
FIG. 1. The rational design of new nanostructures for clinical imaging gray
must holistically consider the imaging modality blue, the imaging target
yellow, and the in vivo properties pink. This differs from the design of
contrast agents for nonhuman subjects green, nonspecific agents purple,
or targeted drug delivery vehicles orange that typically only have to consider two of these three factors.
in vivo properties i.e., its pharmacokinetics, pharmacodynamics, and biodistribution.3639 There are also specific imaging modality-related accessibility issues that must be considered, e.g., the critical condition that relaxation-enhancing
MRI contrast agents have access to water.
Target accessibility must not only consider spatial accessibility, but how long the agent can circulate in the patient to
reach its target before being cleared from the system. The
nanostructures need to be stable enough to circulate in the
blood long enough to localize to disease sites while retaining
their payload/targeting ligands e.g., at least 2 to 3 h are
required for all blood to complete passage through a remote
vascular bed40. Additionally, the nanostructures must be designed to take into account possible elimination by the
mononuclear phagocytic system MPS. This can happen
within a few minutes and minimizes the quantity of nanostructures passing by the target site and, if actively labeled,
significantly decreases their interactions with target antigens.
To avoid MPS elimination the nanostructure surface is typically cloaked, frequently with a hydrophilic polymer e.g.,
polyethylene glycol PEG38,39,41,42 with low immunogenicity and antigenicity.
In opposition to the strategy of avoiding MPS elimination,
nanostructures can be designed for maximal uptake by macrophages in vivo to track their fate for direct and indirect
imaging of disease. For example, certain dextran-coated,
long-circulating, superparamagnetic iron oxide SPIO nanoparticles are preferentially taken up by macrophage cells, and
these cell-encapsulated long-lived entities can thus passively target pathological inflammatory processes for MR
imaging of atherosclerotic plaques.43 However, for effective
in vivo use, the degree to which only target macrophages are
labeled must be established for their use in this capacity.
Alternatively, cell tracking the passive ex vivo labeling
4477
4477
clinical utility. This motivates researchers to either synthesize new agents with only incremental modifications to
clinically accepted technologies or to partially develop
high-risk/high-reward-type agents without rigorous testing or
specific optimization for clinical use. This current paradigm
for agent development is inefficient, expensive, leads to duplication of effort, and has a disproportionate reliance on
luck and/or an ab initio, perfectly defined imaging/biology
problem. This suggests that new, more general approaches to
the design of functional nanostructures are needed.
III. TOWARDS NEW FUNCTIONAL
NANOSTRUCTURES
In our opinion, the next decade will see the rational design of a new generation of nanostructures for medical imaging progressing towards the inclusion of added functionality, with a focus on effective treatment and improved patient
outcome resulting from more sensitive and specific detection
of disease. Already, the role of nanostructures in medical
imaging is expanding toward this goal, from simple detection
of disease to characterization/diagnosis of disease, local application of therapy, real-time assessment of response to
therapy, and as a tool in the drug discovery and development
process.11,4952 The continuing evolution of current nanostructures towards multifunctional, nanoscale medical devices has been explicitly and implicitly anticipated in the
recent literature.11,16,18,26,27,29,5358
We propose that new functional nanostructure designs
will move away from the current paradigm of relatively
simple, spherical nanostructures encapsulating imaging
agents and/or targeting diseased tissues by size-specific or
mono-ligand attachment. In this section, concepts that can
facilitate the development of novel, multifunctional nanostructures will be briefly described, along with proposals on
how new nanostructures could fulfill these functions. These
concepts are classified into five main directions, which are
outlined below: a Modular nanostructures with additive
functionality, b cooperative nanostructures with synergistic
functionality, c nanostructures activated by their in vivo
environment, d nanostructures activated by sources outside
the patient, and e novel, nonspherical nanostructures and
components.
III.A. Evolution of modular nanostructures
with additive functionality
The most obvious tactic for the evolution of multifunctional nanostructures that will permit multiple imaging
and/or therapy applications is a linear add-on approach,
where individual components i.e., imaging agents and/or
therapy agents are combined to form a single multifunctional construct.10,59,60
This add-on approach has been explored in preclinical,
multimodal imaging applications. Numerous multimodal
nanostructures have been proposed, including many dualmodal nanostructures e.g., MRI/SPECT,15,61 MRI/PET,61
MRI/optical,20,6264 and CT/MRI65, and a few tri-modal reporters e.g., for PET, MRI, and fluorescence imaging,66 and
4478
4478
nanostructures can carry contrast agents for virtually all modalities based on this principle.15,17 Also, the structure of
nanoscale constructs may accommodate payloads of mixed
molecular or particulate agents within their volumes or on
their surfaces.
Multifunctional nanostructures comprised of truly modular components will require significant technical advancements, particularly in the area of coatings to integrate different modular components. However, technical progress
may be fast because this approach has already proved to be a
valuable tool for the early, preclinical characterization of
new nanostructures. Not only will this lead to faster assessment and iteration of clinically useful nanostructures at an
early stage of development, new common platform technologies and strategies can thus be developed for the future effective integration of disparate modular components for
clinical imaging. For example, many new imaging nanostructures are being designed to contain an optical marker for
preclinical cross-validation.20,6264 The high sensitivity and
subcellular spatial resolution of optical agents can be used to
assess different targeting strategies in vitro, without altering
the overall properties of the nanostructure. Useful information about the agent may then be obtained prior to moving on
to in vivo models. In the same manner, other validation molecules e.g., those for histopathology60 may be integrated
into early-stage nanostructures to confirm the behavior and
pharmacokinetics of preclinical agents for faster translation
into the clinic.
In general, designing multimodal and multifunctional
nanostructures will remain challenging compared to designing unimodal or unifunctional nanostructures. The integration of two or more different agents within the nanostructure
will add a level of complexity to their synthesis, will decrease the loading capability of each individual agent, and
may change the overall nanostructures pharmacokinetics
and stability. To counter this, nanostructures that provide intrinsic contrast in more than one imaging modality can be
exploited. For example, Gd-chelates can provide contrast in
both MR and CT.73 Perfluorocarbon droplets and their derivatives have been used to provide contrast in CT,7476 US,77
and/or MR7880 imaging, and have also been demonstrated as
drug delivery vehicles81 and as radiosensitizers.82
III.B. Evolution of cooperative nanostructures systems
with synergistic functionality
4479
tive to normal tissue such that the contrast agents can accumulate disproportionately i.e., nonlinearly in the disease
site, or result in a spatial distribution of individual nanoscale
constructs that will otherwise provide signal amplification.
Such signal amplification is significant in imaging applications where increasing signal to noise can decrease the lower
limit for detection currently on the order of hundreds of
millions of cells, as well as in pharmaceutical delivery
where any advantage in therapeutic ratio may play a role in
improved patient outcome.
The simplest example of nonlinear, preferential accumulation of nanostructures is the localization that occurs via the
EPR effect at disease sites with leaky pathologies. In addition to size-dependent accumulation, further nonlinear accumulation may occur by modifications to the nanostructures
surface. This is the underlying concept behind enhancing
size-dependent nanostructure localization in diseased tissue
through active targeting, or the simple attachment of targeting molecules to the nanostructures surface for further imaging or therapeutic amplification.13,16,21,2527 In opposition
to the concept of preferential accumulation at disease sites,
the nanostructures surface may be tailored to assist its clearance and/or elimination from nontarget tissue to reduce background noise and toxicity.8385
More complex strategies for nonlinear accumulation are
possible. This concept is particularly beneficial for the preferential discrimination of disease sites from normal tissues
that express the same biomarkers of disease but at different
levels. The large surface area of the nanostructure compared
to targeting ligands which range from 1 to 20 nm in
size potentially allows the attachment of multiple targeting
ligands. This may facilitate multivalent binding to cell surface receptors such that collections of low-affinity ligands
can have very high effective affinities.8688 This not only
can greatly expand the range of useful, active targeting
ligands, but may allow nanostructures to be conjugated to
multiple weak-binding ligands for disease-specific biomarkers such that they would bind only to sites expressing abnormally high levels of the biomarker, but not at normal, lower
levels. Further enhancement of preferential targeting may be
accomplished by using several sets of nanostructures, each
labeled with weak-binding ligands for different biomarkers
expressed by a disease. A similar strategy, but more technically challenging, would be to use several different biomarkers on the same nanostructure such that disease targets that
express relative proportions of a series of agents will result in
preferential nanostructure accumulation.
The accumulation of nanostructures at a disease site may
be augmented through the use of a series of different sets of
targeted nanostructures. After saturating the disease site with
a first set of targeted nanostructures, it may be possible to
add a second, labeled nanostructure that could attach only to
multiple nanostructures of the first kind, and so on, such that
imaging and/or therapeutic nanostructures could preferentially, and controllably, accumulate at the disease site. In order to prevent nonspecific, uncontrolled accumulation, which
could lead to thrombosis and/or high background imaging
signals, each type of nanostructure must be fully cleared
Medical Physics, Vol. 35, No. 10, October 2008
4479
4480
volume of the nanostructure. They can also have a significantly longer half-life to accumulate and localize in greater
numbers in diseased tissue.
The most straightforward type of environmentally activatable nanostructures simply dissolve and/or aggregate in certain in vivo environments. In fact, the controlled, environmentally dependent degradation of nanostructures into their
molecular components in vivo has been the basis for pharmaceutical delivery of drugs for decades.96,97 More recently,
pH- and temperature-responsive polymeric nanostructures
for potential imaging and drug delivery applications have
been reported in the preclinical research literature.98103
Simple nanostructures can also aggregate in certain in vivo
environments resulting in a significant change in imaging
signal. For example, preclinical iron oxide-based nanostructures for MRI have been shown to undergo reversible clustering in the presence of enzymes, chemical compounds,104
or analytes e.g., glucose105 or calcium106.
One intriguing concept that has not been extensively explored in medical imaging is the controlled in vivo assembly
of nanostructures at the target site. Many nanostructures are
composed of atoms or molecules that assemble together via
intermolecular bonding using weak chemical bonds. Since
nanostructures have large surface-area to mass ratios, surface
effects e.g., intermolecular bonding and surface-dominated
reactions e.g., surface oxidation, saturation of dangling surface bonds are strongly enhanced in nanoscale constructs.
For example, the weak noncovalent forces between molecular components that dominant at the nanoscale i.e., the constructs stability are highly sensitive to slight changes in
their surrounding environmental conditions e.g., temperature, pH107109.
These environmentally sensitive surface properties can be
utilized to modify and/or assemble/disassemble structures
under appropriate conditions in situ, as a function of their
environmental surroundings in a simplified mimicry of biological self-assembly processes i.e., biomimetics.40,110115
Transient, metastable nanostructures can be constructed from
biological or exogenous molecules that will self-assemble
in vivo for short periods of time, depending on the local
environment or external application of energy. This concept
could be expanded further such that nanoscale reaction
vessels116118 for the fabrication of contrast nanostructures
could be assembled within the biological system. The use of
smaller molecular components in vivo that can assemble directly at the target site may allow larger nanostructures to be
built at the target without prior elimination by the MPS.
A simple example of in vivo assembly of a larger structure from smaller nanostructure components can be found in
the preclinical literature, where nanostructures i.e., perfluorocarbon nanodroplets and drug-loaded micelles were injected into an animal model. After injection, the nanostructures underwent temperature-induced coalescence and
conversion to form micron-scale gas-filled microbubbles.119
Further, it was reported that microbubbles created in vivo
not only acted as effective acoustic scatterers for US imaging, they could also be disrupted by US to enhance the delivery of their encapusulated therapeutic cargo.119
Medical Physics, Vol. 35, No. 10, October 2008
4480
The challenges associated with making complex, environmentally activatable contrast nanostructures are that such
nanostructures and their molecular components are currently difficult to synthesize and design, and significant barriers in the complex biological environment of the human
body must be overcome before reaching the target site. One
possible approach to shielding the nanoscale components
from the biological environment is to use a biological entity
as a delivery vehicle. Already, modified lipoproteins120 and
cells have been used as carriers for contrast nanostructures,
and the tracking of stem cells, cancer cells, and macrophages
have been demonstrated in vivo.4345
For environmentally activatable nanostructures to be designed and utilized, and for the considerable technical challenges of synthesizing the agents themselves to be overcome,
it is critical to understand how the contrast signal correlates
to the physical changes in the nanostructure i.e., its degradation and/or self-assembly, how these structural changes
are related to environmental conditions e.g., hypoxia/
oxygenation, pH, and temperature, and how these changes
correlate to disease pathologies.
III.D. Evolution of multifunctional cross-modal
nanostructures that may be activated by devices
external to the patient
New types of nanostructures that can be remotely activated by an energy source external to the patient are also
under investigation. External triggering from energy sources
using different imaging modalities may localize the agent to
its target site, excite the agent to emit a detectable signal
above background noise, and/or produce a local therapeutic
effect.
Externally applied US energy can push contrast nanostructures in the vasculature towards the target site using
radiation force, to increase local binding121 or to cause individual particles to attract each other and coalesce.122 This
same concept may allow US to be used to improve the localization of other i.e., non-US imaging and therapy nanostructures. This type of controlled localization of selected
structures external to the patient may also eventually facilitate the directed self-assembly of larger constructs in vivo at
a target site.
In another example of localization, externally applied
magnetic fields have been used to control and track magnetic
beads using magnetic field gradients to increase localization/
binding to target sites in preclinical systems.123126 The concept of enhanced localization of drugs using this method may
be envisioned, as magnetic particles have been coupled with
pharmaceutical agents124 and therapeutic radiolabeled
molecules.126
Imaging modalities have been used to activate an exogenous contrast nanostructure in vivo from outside the body.
For example, US can convert contrast nanostructures e.g.,
perfluorocarbon emulsions to gas bubbles, thereby significantly increasing their acoustic contrast127 and signal to noise
such that single bubbles may be imaged in vivo. The energy
of the imaging system can also be applied to nanostructures
4481
4481
4482
effect has been the key to developing imaging and pharmaceutical nanostructures for cancer detection and therapy.
The emergence of new imaging modalities should play a
role in propelling the development of new complementary
contrast agents and increase the functionality of existing
nanostructures. One example gaining in popularity is transcutaneous optical imaging, which has led to the development
of biocompatible molecular near-infrared probes.148 In addition to current fluorescence and luminescence methods, other
aspects of optical technology including absorption, emission, and scattering techniques will require the optimization
of new contrast agents to complement these different contrast
mechanisms.
The main challenge in developing new types of nanostructures for functional imaging is that they must truly be
rationally designed for them to be successful. A thorough
understanding of the imaging and disease parameters is necessary, and the dependence on variations in size, composition, and morphology should be established beforehand. The
design of new nanostructure must be balanced with practical
issues of cost and yield, which will depend greatly on the
synthesis method.
IV. STRATEGIES FOR MOVING FORWARD
Despite the innumerable nanostructures outlined in the
preclinical research literature, the translation of new nanostructures into humans is in the early stages. For example, in
2006, the only marketed nanoscale contrast agents were three
superparamagnetic iron oxide nanoparticle contrast agents
for MRI.69 To facilitate the rapid progression of new, functional nanostructures into the clinic, we must address three
main challenges: a Improving the clarity of both the desired
nanostructure properties and the challenges needed to be
overcome for their successful implementation, b determining better methods to test and validate the new nanostructures early in their development, and c developing accepted
and established strategies for the efficient translation of new
nanostructures from the lab bench into the clinic.
IV.A. Improved clarity of desired properties and
existing challenges
The development of new functional contrast agents requires the convergence of highly specialized and disparate
fields. In addition to overcoming the biological issues effective delivery to the target, biocompatibility, toxicity and fabrication issues synthesis, yield, quality assurance as outlined earlier in this review, it is also necessary to overcome
medical imaging issues to obtain sufficient contrast for imaging.
Through collaboration with physicians, biologists, and
chemists, medical physicists must determine the appropriate
design parameters, such as minimum required loading, and
ideal agent and imaging timepoints, in parallel with the spatial resolution required for disease diagnosis. For contrast
agent design and determination of minimum detection limits,
the expected number, location, and distribution of the target
should be known. The overall suitability of the particular
Medical Physics, Vol. 35, No. 10, October 2008
4482
imaging modality also depends on the expected target distribution within the patiente.g., whole body tomographic
imaging can be done with CT, MRI, and nuclear imaging,
but is not commonly undertaken with US or optical imaging.
For combined detection and therapy, the imaging properties
must be weighed with the desired effect of the therapeutic
agent, as determined by its degradation rate, concentration,
and effective target delivery. All these factors must be considered for the efficient development of new nanostructures
for medical imaging, and will facilitate the design of new
contrast agents and imaging techniques to allow not only the
agents detection but also their quantification, allowing specific information about the changes in expression levels of
the target to be obtained.
The wide knowledge gap between the disparate fields involved in the design of new functional nanostructures must
be addressed. Critical bottlenecks in nanostructure agent development are often not elucidated preventing progress in
creating suitable nanostructures for medical imaging. Recently, many institutes and government agencies have recognized the importance of communication within this interdisciplinary ensemble of highly specialized fields and have
implemented interdisciplinary teams to increase the efficiency of their interactions.57,149,150
Another simple way to link the scientific community is to
utilize the massive information sharing capability of the internet to develop a database of core parameters that identify
different types of imaging and therapeutic nanostructures,
their correlative compositions, and their imaging and in vivo
properties. Recently, the National Center for Biotechnology
Information NCBI at the National Institutes of Health
NIH implemented such a freely accessible online database
MICAD151 to provide the scientific community working
with in vivo molecular imaging and contrast agents with information aimed to foster research and development. A similar database focusing on nanoscale agents should be developed, supplemented by correlative databases that identify the
parameters desired from an imaging perspective e.g., ideal
type/number/distribution of molecular or atomic units required per unit, size range, amount of agent required, special
considerations/parameters, a biological perspective e.g.,
ideal disease targets, targeting ligands/biomarkers, delivery
paths, safety data, and a commercial/market perspective
e.g., the desired yield, purity and final application/market
demand and size. In addition to being freely accessible,
these databases should be linked, cross-searchable, and able
to accommodate dynamic adaptation by the users. Although
some specific fabrication protocols may be restricted due to
patenting and commercialization concerns, the protocols and
procedures employed for characterization of the agents as
well as their properties should be listed, which will assist in
establishing baseline standards for development of new
nanostructure agents. This massive collaborative effort
should help differentiate which nanostructures are in the
early, mid, and late stages of development; identify major
knowledge gaps and the likelihood of the successful translation of various agents to the clinic; and help scientists to
4483
converge toward the most optimal nanostructure design parameters to yield the greatest patient benefit.
IV.B. Development of early-stage validation and
characterization tools specific for nanostructure
imaging and therapy applications
First, the development of common standards and validation tools for functional nanostructure assessment will be
necessary to streamline and adequately evaluate the thousands of nanostructures under investigation for imaging and
therapy applications reported in the literature. Currently,
even agreement on what constitutes agent size has not been
standardized e.g., hydrodynamic radius versus dry radius, and something as simple as the adoption of common
units to quantify size would help alleviate the confusion between chemists who measure size using atomic mass Daltons and physicists who measure linear dimension nanometers. Also, it would be beneficial to establish goldstandard protocols for the assessment of toxicity/safety and
bioconjugation, and for allowing different agents and numerous and occasionally contradictory protocols in the literature to be compared, while reducing the abundance of research nanostructures to those that have met a known,
accepted standard. In addition, the development of validation
tools specifically for nanostructure imaging agents would be
useful. Currently, most researchers cannot image nanostructures on a subcellular scale without an integrated optical
marker. The development of a suite of subcellular and other
high-resolution imaging tools that mimic clinical systems
may allow early assessment of nanostructures, which could
lead to faster iteration of nanostructure design in the early
developmental stage.
Second, it would be very useful if common platform technologies could be developed such that the targeting ligands,
bioconjugation strategies, surface, interfacial, and capping
layers could be tested independently of the imaging construct. In this way, it may be possible to develop a consistent
description of the behavior of nanostructures in biological
systems as a function of nanostructure size, structure, and
material composition. The ability to tune the surfaces to be
efficiently capped by various targeting/shielding ligands
would also assist in differentiating the properties of the nanostructure from the properties of their surfaces e.g., structure
vs. charge. Current challenges such as hydrodynamic size
effects, pH stability of the composite structures, and reproducibility biological versus structural issues may be addressed using universal nanoscale constructs. This can lead
to predictive models of nanostructure systems, and further to
the engineering of specific nanostructures tailored to biological applications.
Already, the concept of using standardized, validated
nano-based imaging agents for the real-time assessment of
the efficacy of therapeutic regimens as a complement to, or a
substitute for, conventional end point analysis has been
proposed.26 This application can assist in the design and advancement of standardized, nanostructure-based validation
tools.
Medical Physics, Vol. 35, No. 10, October 2008
4483
4484
19
4484
4485
47
4485
47294749 2006.
S. B. Yu and A. D. Watson, Metal-based x-ray contrast media, Chem.
Rev. Washington, D.C. 999, 23532378 1999.
74
R. F. Mattrey, D. M. Long, F. Multer, R. Mitten, and C. B. Higgins,
Perfluoroctylbromide: A reticuloendothelial-specific and tumor-imaging
agent for computed tomography, Radiology 1453, 755758 1982.
75
D. M. Long, Jr., E. C. Lasser, C. M. Sharts, F. K. Multer, and M. Nielsen,
Experiments with radiopaque perfluorocarbon emulsions for selective
opacification of organs and total body angiography, Invest. Radiol.
153, 242247 1980.
76
D. M. Long, F. K. Multer, A. G. Greenburg, G. W. Peskin, E. C. Lasser,
W. G. Wickham, and C. M. Sharts, Tumor imaging with x-rays using
macrophage uptake of radiopaque fluorocarbon emulsions, Surgery St.
Louis 841, 104112 1978.
77
C. W. Hughes, R. W. Williams, M. Bradley, and G. H. Irvine, Ultrasound
monitoring of distraction osteogenesis, Br. J. Oral Maxillofac Surg.
414, 256258 2003.
78
A. M. Morawski, G. A. Lanza, and S. A. Wickline, Targeted contrast
agents for magnetic resonance imaging and ultrasound, Curr. Opin. Biotechnol. 161, 8992 2005.
79
G. M. Lanza, R. Lamerichs, S. Caruthers, and S. A. Wickline, Molecular
Imaging in MR with targeted paramagnetic nanoparticles, Med. Mundi
471, 3439 2003.
80
S. D. Caruthers, A. M. Neubauer, F. D. Hockett, R. Lamerichs, P. M.
Winter, M. J. Scott, P. J. Gaffney, S. A. Wickline, and G. M. Lanza, In
vitro demonstration using 19F magnetic resonance to augment molecular
imaging with paramagnetic perfluorocarbon nanoparticles at 1.5 Tesla,
Invest. Radiol. 413, 305312 2006.
81
Y. B. Yu, Fluorocarbon nanoparticles as multifunctional drug delivery
vehicles, J. Drug Target. 1410, 663669 2006.
82
S. Rockwell, M. Kelley, C. G. Irvin, C. S. Hughes, E. Porter, H. Yabuki,
and J. J. Fischer, Modulation of tumor oxygenation and radiosensitivity
by a perfluorooctylbromide emulsion, Radiother. Oncol. 222, 9298
1991.
83
C. Wilhelm, C. Billotey, J. Roger, J. N. Pons, J. C. Bacri, and F. Gazeau,
Intracellular uptake of anionic superparamagnetic nanoparticles as a
function of their surface coating, Biomaterials 246, 10011011 2003.
84
J. Kreuter, The influence of coatings with surfactants on the body distribution of nanoparticles after intravenous injection to rats, Clini. Materi.
1314, 131134 1993.
85
V. Labhasetwar, C. Song, W. Humphrey, R. Shebuski, and R. J. Levy,
Arterial uptake of biodegradable nanoparticles: Effect of surface modifications, J. Pharm. Sci. 8710, 12291234 1998.
86
C. B. Carlson, P. Mowery, R. M. Owen, E. C. Dykhuizen, and L. L.
Kiessling, Selective tumor cell targeting using low-affinity, multivalent
interactions, ACS Chem. Biol. 22, 119127 2007.
87
S. Hong, P. R. Leroueil, I. J. Majoros, B. G. Orr, J. R. Baker, Jr., and M.
M. Banaszak Holl, The binding avidity of a nanoparticle-based multivalent targeted drug delivery platform, Chem. Biol. 141, 107115 2007.
88
X. Montet, M. Funovics, K. Montet-Abou, R. Weissleder, and L. Josephson, Multivalent effects of RGD peptides obtained by nanoparticle display, J. Med. Chem. 4920, 60876093 2006.
89
A. Tanimoto, K. Oshio, M. Suematsu, D. Pouliquen, and D. D. Stark,
Relaxation effects of clustered particles, J. Magn. Reson Imaging 141,
7277 2001.
90
O. Couture, P. D. Bevan, E. Cherin, K. Cheung, P. N. Burns, and F. S.
Foster, Investigating perfluorohexane particles with high-frequency ultrasound, Ultrasound Med. Biol. 321, 7382 2006.
91
P. Caravan, Strategies for increasing the sensitivity of gadolinium based
MRI contrast agents, Chem. Soc. Rev. 356, 512523 2006.
92
U. I. Tromsdorf, N. C. Bigall, M. G. Kaul, O. T. Bruns, M. S. Nikolic, B.
Mollwitz, R. A. Sperling, R. Reimer, H. Hohenberg, W. J. Parak, S.
Forster, U. Beisiegel, G. Adam, and H. Weller, Size and surface effects
on the MRI relaxivity of manganese ferrite nanoparticle contrast agents,
Nano Lett. 78, 24222427 2007.
93
A. Y. Louie, M. M. Huber, E. T. Ahrens, U. Rothbacher, R. Moats, R. E.
Jacobs, S. E. Fraser, and T. J. Meade, In vivo visualization of gene
expression using magnetic resonance imaging, Nat. Biotechnol. 183,
321325 2000.
94
J. Rao, A. Dragulescu-Andrasi, and H. Yao, Fluorescence imaging in
vivo: Recent advances, Curr. Opin. Biotechnol. 181, 1725 2007.
95
S. Kumar and R. Richards-Kortum, Optical molecular imaging agents
for cancer diagnostics and therapeutics, Small 11, 2330 2006.
73
4486
4486
4487
4487
Received 27 October 2007; revised 29 July 2008; accepted for publication 29 July 2008;
published 17 September 2008
The nuclear magnetic resonance phenomenon has given rise to both magnetic resonance imaging,
which yields morphologic data, and magnetic resonance spectroscopy MRS, which yields chemical data. In humans these data are derived principally from the resonances of the hydrogen nucleus
in the low molecular weight compounds in the body. Hydrogen MRS has become a routinely used
clinical tool in the brain, prostate, and breast. Other nuclei also demonstrate this phenomenon but
each of these comes with additional difficulties, including low abundance, low sensitivity, and/or
low chemical concentrations. The future of MRS includes a drive to higher main magnetic field
strengths and new methods to create 45 orders of magnitude greater signal. The future of MRS is
bright, but in the United States it is endangered by overuse and misuse driven by the advent of
reimbursement. 2008 American Association of Physicists in Medicine.
DOI: 10.1118/1.2975225
I. INTRODUCTION
I.A. Comparing magnetic resonance imaging and
magnetic resonance spectroscopy
The NMR effect is exceedingly small. Of all of the hydrogen nuclei in the human body, only 1 in 105 is polarized
when immersed in a field of 1.5 T. The signal which can be
derived from any NMR experiment MRI or MRS is first
dependent on this polarization. In all but the most recent
0094-2405/2008/3510/4530/15/$23.00
4530
4531
4531
FIG. 1. Localizing MRI and accompanying 1H MRS spectrum from a 0.56 cm3 voxel in the left putamen. Data acquired at 0.5 T with short echo time elliptical
excitation chemical shift imaging. Acquired as part of a repeatability study. Data shown in b was from the second session when the volunteer was inebriated.
Triplet resonance of ethanol marked ETOH.
Improving MRS data is usually accomplished by improving the signal to noise ratio of the data and by increasing the
separation between resonances. Signal to noise ratio in MR
spectroscopy is a linear function of field strength and is
given by5
SNR =
B0KNNEXeTe/T21 eTr/T1
lwbw
4532
4532
FIG. 3. Ethanol phantom 1H spectra demonstrating change of chemical shift with main magnetic field B0 strength. In a and c, spectra are displayed with
chemical shift in hertz with water resonance set to 0 Hz. In b and d, spectra are displayed in ppm showing that the centers of each multiplet are at the same
offset in ppm. Frequency splitting of the multiplets in d appears larger than in b because J coupling is invariant to change in field strength, whereas
chemical shift is linear with B0.
These effects are referred to as homo- and heteronuclear coupling, respectively. Also known as J coupling, it causes the
resonance to split into two or more resonances, depending on
the number of other nuclei involved in the coupling and the
magnitude of the coupling, as shown in Fig. 4. The total
amount of energy in the resonance is split between the individual resonances with an energy distribution which is dependent on the quantum states of the spins in the coupled
nuclei.6 Two coupled spins form a doublet with two resonances of equal amplitude 1:1 and three coupled spins form
a 1:2:1 pattern, while four create a 1:3:3:1 pattern. The magnitude of the coupling is independent of field strength, and is
measured in hertz, not ppm. The appearance of coupled resonances is a function of the ratio of / J, which is a function
of field strength. As B0 approaches zero, the coupled resonances degenerate into a singlet as the ratio / J approaches
zero.7 At very high values of B0, the individual resonances in
the multiplet may separate sufficiently to be individually deMedical Physics, Vol. 35, No. 10, October 2008
4533
4533
FIG. 4. Homonuclear and heteronuclear spin J coupling in a and b, respectively, showing the paths of coupling. Note that the coupling is much smaller
for the homonuclear case. Removal of heteronuclear splittings by proton decoupling shown in top spectrum on the right in b. The area and amplitude of the
resulting resonance after decoupling is greater than the coupled case as energy split between resonances in the multiplet are summed when the J coupling is
removed.
New techniques in clinical MRI or MRS are first developed using phantom and animal models in preclinical, small
bore systems, followed by evolution of whole-body magnet
technology and proof of safety and efficacy through human
clinical trials. One of the first methods proposed for MR
imaging by Mansfield in 1977 was echo planar imaging
where the spin system is excited and the entire k-space plane
is acquired by a string of phase and frequency encoding graMedical Physics, Vol. 35, No. 10, October 2008
4534
4534
FIG. 5. Phantom study of the change of resonance appearance with field strength. The creatine resonances are singlets which do not change appearance with
field strength while coupled resonances of glutamate and taurine are strongly affected. Phantom composed of 20 mM each creatine, glutamate, and taurine in
water, titrated to physiologic pH.
FIG. 6. Data acquired at 1.5 T in a brain mimicking phantom at constant repetition time TR and multiple echo times TE. Note that the phase of the doublet
of lactate at 1.3 ppm changes phase with echo time. This forms the basis of J-coupled spectroscopy. Also it can be seen that each of the metabolites changes
amplitude with TE based on the T2 of that resonance. Therefore, observation of some metabolites such as glutamate requires short TE times.
Medical Physics, Vol. 35, No. 10, October 2008
4535
lesions in patients were due to histological changes associated with those lesions14 Since that time, 1H MRS has moved
into routine use in patients.
II. THE STATE OF THE ART
II.A. 1H MR Spectroscopy
4535
4536
4536
FIG. 7. Sum of spectra in a, from a single slice CSI acquisition in a patient with a newly appearing lesion shows a characteristic pattern for neoplasm
increased choline and decreased NAA. Plotting several spectra as shown in b demonstrates the necrotic core of the lesion with changing neoplastic features.
The use of this spatial data provides the actual tissue diagnosis, glioblastoma. Data acquired at 0.5 T using a short echo time CSI sequence.
Medical Physics, Vol. 35, No. 10, October 2008
4537
4537
Despite the ability of CSI to acquire data from many voxels simultaneously, single voxel MRS methods retain significant diagnostic utility. Some smaller lesions in areas of the
brain which may be surrounded by air, bone, or adipose tissue are not amenable to exploration by CSI methods due to
contamination from adjacent voxels which may sample tissue outside of parenchyma. For example, the lesion shown in
Fig. 8 could only be sampled with a single voxel technique.
Another compelling reason to acquire data by the single
Medical Physics, Vol. 35, No. 10, October 2008
4538
4538
FIG. 9. Images from two patients with similar lesions referred for 1H MRS
of newly appearing lesions. Images on the right have a false color overlay of
the ratio of choline to NAA. Lesion in the upper patient clearly demonstrates
neoplasm Cho/ NAA 1 while the image below shows that the lesion is
not neoplastic.
4539
FIG. 10. Chemical shift image of glutamate combined with the postcontrast
T1-weighted image of a patient with a glioblastoma. Also on this image is
the 98% dose contour line of the radiation therapy plan shown as the thin
red line. The site of eventual recurrence of this tumor was posterior to this
lesion and is predicted by the red elevated glutamate area Ref. 63.
4539
is decoupling. It requires yet more hardware and is exquisitely sensitive to artifact. Proton decoupling of the 13C spectrum of ethanol is shown in Fig. 4b.
Phosphorus spectroscopy detects adenosine triphosphate
ATP, phosphocreatine PCr, inorganic phosphate Pi, the
phosphomono- PME, and phosphodiesters. Studies of brain
have shown alterations of phospholipid metabolism in psychiatric disorders.39 Cardiac function can also be studied by
31
P spectroscopy.40 Dynamic studies of muscle are presently
being used to assess muscle recovery during isometric or
dynamic exercise as shown in Fig. 11.
Glycogen in muscle has been studied in humans with the
use of the 13C.41 Glucose metabolism and its interaction with
neuronal function has been studied in brain using 13C labeled
glucose.42 Similar studies are giving insight into neuronastrocyte trafficking in glutamate and glutamine and the diseases associated with the dysregulation of this process.43
Other nuclei which are not otherwise present in appreciable abundance in the body have been the subject of MRS
studies. These include lithium in the brains of bipolar patients and fluorine in antipsychotic drugs in brain and chemotherapeutic drugs in liver.44
Given the additional hardware required and the additional
complications of acquisition, no X nucleus exams are in
widespread clinical use.
4540
4540
FIG. 11. 31P MRS spectrum from the gastrocnemious muscle of a patient in an isometric contraction apparatus. 15 s contraction demonstrates the decrease of
PCr heavy arrow and the increase of Pi thin arrow.
V. FUTURE DEVELOPMENTS IN MR
SPECTROSCOPY
The brain will continue to be one of the most studied
organs by MRS. The ability to extract functional data noninvasively continues to be the driving force behind MRS. This
is likely to remain so in the future. Several benefits accrue to
1
H MRS from increasing field strength. These include the
ability to separate resonances that are too closely spaced at
lower field strengths, most notably 1.5 T. The increased signal to noise and chemical shift dispersion of increased field
strength also improves metabolite editing sequences. An example is the detection of the inhibitory neurotransmitter
GABA which nominally is obscured by the creatine
resonance.48 Increased SNR from operation at high field and
the use of phased array surface coils allow data acquisition
with either shorter total scan times or with higher spatial
resolution. Similar phase undersampling strategies used by
MRI for scan time reduction can also be applied to MRS.49
One further benefit of increased chemical shift dispersion is
the ease with which chemical shift saturation water saturaMedical Physics, Vol. 35, No. 10, October 2008
4541
4541
FIG. 12. Spectra from a 3.4 cm3 voxel acquired in the occipital lobe of a healthy volunteer acquired under identical conditions at three different magnetic field
strengths. Note the increased amplitude of the glutamate/glutamine resonance Glx at 0.5 T relative to 1.5 T. While the individual resonances in the multiplet
start to become visible at 3 T, they are still clearly overlapped. The prominence of the myoinositol resonance also decreases with increasing field strength.
terface above the sinuses and the imperfections of field homogeneity, both of which increase with field strength, cause
losses in the SNR. Third, many of the resonances which
overlap at lower field strengths continue to do so even at
field strengths above 7 T. As the MRS field matures, a more
balanced view may become predominant. Many disease
states will benefit from observation at lower field strengths.
Reduced linewidths and the collapse of multiplets into pseudosinglets favor operation at field strengths as low as
0.5 T.52 Figure 12 illustrates the change of spectral appearance with field strength in the same voxel of a volunteer
acquired at three different field strengths. Note that the methylene resonances of glutamate and glutamine at 2.3 ppm
increase in amplitude at 0.5 T. A similar effect is shown in
the myoinositol resonance at 3.56 ppm. Even taurine benefits, as was shown in Fig. 5. Magnet designs intended for
obese and/or claustrophobic patients with field strengths beMedical Physics, Vol. 35, No. 10, October 2008
4542
FIG. 13. Hyperpolarized gas images from the lungs of a healthy subject
comparing 3He to 129Xe. Images courtesy of Dr. John Mugler, University of
Virginia.
the body in ionic form. Having only one moiety, sodium can
be imaged using high speed MRI methods. Phosphorus-31
has 100% isotopic abundance unlike 13C which is only 1.1%
abundant. All of the X nuclei observed with natural polarization will benefit from higher field strength magnets. The increased field strength is needed as the intrinsic signals from a
normally polarized 13C or 31P nucleus are 0.016 and 0.066
that of a hydrogen nucleus. Additional transmit channels can
be used to eliminate heteronuclear J couplings of the X nuclei with 1H as shown in Fig. 4. The signal intensity of the X
nucleus resonance then increases as decoupling removes the
splittings. An additional effect to gain signal, utilizing the
same hardware, is nuclear Overhauser enhancement NOE
which can create an increase in the polarization. Decoupling
combined with NOE can provide two to fourfold signal
increases.54 Equipping a MRI system with both the additional transmit and receive channels, RF coils, and associated
hardware to achieve decoupling can cost a substantial fraction of the cost of the original MRI system. That is a large
investment for what is still a time consuming and difficult
study. There is now a method of increasing the polarization
of the nuclei of interest, called hyperpolarization.55 Helium-3
and xenon-129 can be polarized to 50% or more. This fact is
quite startling when one realizes that the polarization of 1H
in the body at 1.5 T is 1:100 000. The hyperpolarized gases
have T1 relaxation times ex vivo on the order of 10 min.4,56
4542
This extraordinarily long T1 means that the gas can be polarized, stored in a magnetic bottle, and carried to the scanner. Studies of lung function are now being carried out with
3
He and 129Xe as shown in Fig. 13. Xenon has been used for
cerebral perfusion studies in computed tomography where it
changes tissue x-ray attenuation. In MRI, xenon can be used
to study cerebral perfusion as it readily dissolves into blood
through alveolar exchange. The xenon resonance frequency
is sensitive to surrounding tissues. A chemical shift in excess
of 200 ppm is created by proximity to the alveolar surface
and thus can be used to study not just ventilation but lung
function as well.55 The prepolarization of the gas means that
a very high field magnet is not needed to yield signal.
Smaller, more open and patient-friendly magnets will be sufficient to create both imaging conditions and the 1H localization images.
The most exciting new development in spectroscopy is
the hyperpolarization of 13C. Achieved in a different manner
from hyperpolarization of the previously mentioned gases, a
large range of bioavailable chemicals can be created with the
polarized 13C in their structure. To date, two different methods for achieving high levels of polarization in carbon-13
bearing compounds have been proposed. The first is called
dynamic nuclear polarization.57 In this method, the 13C enriched compound in the solid state undergoes microwave irradiation in a 3.3 T magnet while the sample is held at 1 K.
Once polarized, the sample is rapidly dissolved in a suitable
solvent, purified, and injected into the subject who is lying in
the MRI magnet. The second method is polarization transfer
from parahydrogen PHIP in which the polarization from
the parahydrogen is transferred to a 13C containing molecule
through a rhodium catalyst.58 While naturally polarized
carbon-13 labeled glucose has been used to measure the rate
of the tricarboxilic acid cycle in brain,59 the technique requires long acquisition times and high magnetic field
strengths. Hyperpolarization carries the potential to create
metabolic tracers similar in function to those used in positron
emission tomography. Unlike fluorodeoxyglucose FDG, the
tracers created using hyperpolarized 13C are chemically identical to those in the body. In postitron emission tomography
FIG. 14. Location of the image slab in the rat and the corresponding transversal 1H-NMR image. The NMR signal distribution obtained simultaneously from
pyruvate, lactate, and alanine is calculated, and the color images representing the intensity of each metabolite are projected on the anatomical 1H image.
Alanine is most prominent in the skeletal muscle around the spinal cord, whereas the P22 tumor tissue is indicated by the highest signal for lactate. Note the
different scale. Images courtesy of Klaes Golman, Rene int Zandt, Mathilde Lerche, Rikard Pehrson, and Jan Henrik Ardenkjaer-Larsen, Amersham Health
R&D AB Part of GE Healthcare, Malm, Sweden.
Medical Physics, Vol. 35, No. 10, October 2008
4543
4543
VI. CONCLUSIONS
In the past 15 years 1H MR spectroscopy has moved from
the laboratory into routine use for patient care. MR system
manufacturers continue to improve their systems to meet the
stringent demands which MRS places on machine performance. Higher field strength MR systems will improve many
facets of spectral data quality, but bring their own complications, such as a heightened sensitivity to brain iron and a
change in metabolite ratios. Most clinical spectra are still
qualitatively interpreted by the ratio of resonance amplitudes. Changing field strength changes what constitutes normal. These changes need to be measured across patient populations and used in patient data analysis. Improvements in
the analysis and interpretation of spectra, such as time domain, automated fitting methods in the frequency domain,
and feature-based algorithms to classify tissue types, will
address these field strength questions as well as improve the
utility and accessibility of 1H MRS to the clinician. Using 1H
MRS to not only distinguish tissue types but to predict recurrence or premorphologic tumor spread will secure a place
in for 1H MRS in the clinicians retinue of patient management tools.61
Future directions in 1H MRS include smaller voxel volumes which will improve the ability to distinguish healthy
from diseased tissue. Decreasing voxel volumes while maintaining clinically useful acquisition times will require higher
field strength magnets and improved RF coil designs, such as
phased array head coils. Increased use of multiple voxel acquisition methods such as CSI will make MRS less sensitive
to the skills of the operator and clinician in prescribing a
voxel location. A CSI study with small voxels, aided by increased SNR from the use of phased array coils and higher
field strength magnets will make the retrospective analysis of
Medical Physics, Vol. 35, No. 10, October 2008
4544
4544
S. Jordan et al., AHQR 2007 2003.
G. Bielicki et al., NMR Biomed. 17, 60 2004.
49
X. Zhao et al., Magn. Reson. Med. 53, 30 2005.
50
C. H. Meyer et al., Magn. Reson. Med. 15, 287 1990.
51
J. Vymazal et al., J. Neurol. Sci. 134, 19 1995.
52
R. W. Prost et al., Magn. Reson. Med. 37, 615 1997.
53
T. G. Perkins et al., ISMRM, Berlin, Germany, 2007 unpublished.
54
D. W. Klomp et al., Magn. Reson. Med. 55, 271 2006.
55
M. S. Albert et al., Nature London 370, 199 1994.
56
K. Ruppert et al., Magn. Reson. Med. 44, 349 2000.
57
J. H. Ardenkjaer-Larsen et al., Proc. Natl. Acad. Sci. U.S.A. 100, 10158
2003.
58
M. Goldman et al., Magn. Reson. Imaging 23, 153 2005.
59
G. F. Mason et al., J. Neurochem. 100, 73 2007.
60
K. Golman et al., Cancer Res. 66, 10855 2006.
61
E. E. Graves et al., Neurosurgery 46, 319 2000.
62
W. Hollingworth et al., AJNR Am. J. Neuroradiol. 27, 1404 1404.
63
T. Takano et al., Nat. Med. 7, 1010 2001.
48
Received 9 February 2008; revised 24 June 2008; accepted for publication 26 August 2008;
published 14 October 2008
Breast imaging is largely indicated for detection, diagnosis, and clinical management of breast
cancer and for evaluation of the integrity of breast implants. In this work, a prospective view of
techniques for breast cancer detection and diagnosis is provided based on an assessment of current
trends. The potential role of emerging techniques that are under various stages of research and
development is also addressed. It appears that the primary imaging tool for breast cancer screening
in the next decade will be high-resolution, high-contrast, anatomical x-ray imaging with or without
depth information. MRI and ultrasonography will have an increasingly important adjunctive role for
imaging high-risk patients and women with dense breasts. Pilot studies with dedicated breast CT
have demonstrated high-resolution three-dimensional imaging capabilities, but several technological barriers must be overcome before clinical adoption. Radionuclide based imaging techniques and
x-ray imaging with intravenously injected contrast offer substantial potential as a diagnostic tools
and for evaluation of suspicious lesions. Developing optical and electromagnetic imaging techniques hold significant potential for physiologic information and they are likely to be of most value
when integrated with or adjunctively used with techniques that provide anatomic information.
Experimental studies with breast specimens suggest that phase-sensitive x-ray imaging techniques
can provide edge enhancement and contrast improvement but more research is needed to evaluate
their potential role in clinical breast imaging. From the technological perspective, in addition to
improvements within each modality, there is likely to be a trend towards multi-modality systems
that combine anatomic with physiologic information. We are also likely to transition from a standardized screening, where all women undergo the same imaging exam mammography, to selection of a screening modality or modalities based an individual-risk or other classification. 2008
American Association of Physicists in Medicine. DOI: 10.1118/1.2986144
Key words: breast cancer, mammography, digital mammography, tomosynthesis, CT, ultrasound,
MRI, contrast agents, optical imaging
I. INTRODUCTION
II. MAMMOGRAPHY
Mammography is essentially the only widely used imaging modality for breast cancer screening. Various forms of
radiographic imaging of the breast have been used for nearly
one hundred years but mammography with dedicated equipment and technique did not emerge as a screening tool until
the mid-1960s.2 The importance of physical compression of
the breast and the significant association between microcalcifications and breast carcinoma were recognized in the early
1950s by Leborgne.3 While Gershon-Cohen in the United
States and Gros in Europe were strong advocates for breast
cancer screening, the development of a low kVp, high mAs
technique that can be performed in a reproducible manner in
1960 by Egan enabled an organized breast cancer screening
program.4 In 1966, Compagnie Gnrale de Radiologie
CGR, France in collaboration with Gros developed the first
dedicated mammography unit. This unit featured a molybdenum anode with a nominal 0.7-mm focal spot. Subsequently,
several system manufacturers developed dedicated mammography units that facilitated its widespread availability.
Several large randomized clinical trials have shown that
mammography reduces mortality from breast cancer.510
4878
0094-2405/2008/3511/4878/20/$23.00
4878
4879
4879
raphy with screen-film mammography in a screening population demonstrate equivalency for cancer detection,1921 and
digital mammography performed significantly better for preand perimenopausal women younger than 50 years with
dense breasts.22
Current technological approaches for digital mammography that are in clinical use can be broadly classified into
fixed-detector and flexible-detector installations. In a
fixed-detector installation, the dedicated detector is hardwired to the support column of the mammography system
and is not removable. Examples of such fixed-detector installations include indirect conversion hydrogenated amorphous silicon a-Si:H based detectors and direct conversion
amorphous selenium a-Se based detectors. In contrast, a
flexible-detector installation uses the x-ray cassette holder
of a mammography system to house the detector. An example of such a flexible-detector system is computed radiography CR technology. One advantage of the flexibledetector installation is the ease of conversion of an existing
screen-film mammography system to a digital system.
An indirect conversion system uses an intermediary stage,
typically a scintillator to convert the transmitted x-rays to
light photons, followed by detection of the converted light
photons using an optical sensor. The scintillator of choice for
indirect conversion systems is CsI:Tl, which, due to its columnar structure, suppresses lateral light diffusion and,
hence, results in better preservation of spatial resolution. Development of a-Si:H arrays was an important contributor for
adaptation to large field of view full-field imaging.23 Physical characterization indicate a substantial improvement in
DQE 0.55 at zero-spatial frequency characteristics compared to screen-film mammography.18,24
A direct conversion system does not use an intermediary
stage and conversion is made from x-rays to electrons after
interaction in a photoconductive layer, typically a-Se. While
a-Se was used more than four decades ago in xeroradiography and xeromammography, the development of advanced
charge readout methods enabled its adaptation to digital
mammography.25,26 The elimination of the intermediate scintillator layer with direct detection systems allows such systems to achieve high spatial resolution,27 and higher DQE
characteristics 0.54 to 0.64 at zero-spatial frequency compared to screen-film mammography.17 In addition, a slot-scan
photon-counting digital mammography system that uses silicon strip detectors is in clinical use in some countries. This
approach, in addition to providing efficient scatter rejection
due to the slot-scan geometry,28,29 substantially reduces electronic noise contribution to the acquired image resulting in
improved detective quantum efficiency.30
Computed radiography CR uses a photostimulable phosphor plate typically consisting of BaFBr, I : Eu2+ crystals on
a suitable substrate in the form of a portable cassette. When
exposed to x rays, f-centers are created in proportion to x-ray
exposure and remain stable for several hours. This latent
image is read out by stimulating the photostimulable phosphor plate with a laser beam, typically in a raster fashion,
resulting in the emission of light photons in the ultraviolet-
4880
4880
In CR technology, some of the important advances include the development of columnar stimulable phosphor
screens based on CsBr,51 development of linear readout line
scan technology for faster readout of the phosphor,52 and
integration of these approaches.53,54 However, we are not
aware of their transition to digital mammography as yet, but
that could likely occur in the near future.
Another area of substantial interest in detector technology
is the development of energy-resolving large-area photoncounting detectors, which has been challenging. While several approaches for photon-counting detectors have been proposed including scintillation detection using micro-channel
plates55 and gaseous detectors, it appears that semiconductorbased direct-conversion photon-counting systems may be
better suited for medical imaging.56 Some of the semiconductor materials that are considered promising for photoncounting detectors include Si, GaAs, CdTe, and CdZnTe,
typically bonded to a two-dimensional readout with application specific integrated circuits, and are commonly referred
to as hybrid detectors. Several collaborative research projects
to develop hybrid detectors exist,57,58 increasing the prospects for their availability in the next decade.
The advent of digital mammography has prompted reconsideration of several aspects of mammographic imaging that
translated from screen-film imaging. These include anode
material, x-ray spectra, technique factors, and radiation dose.
In addition to the common target-filter combinations of MoMo, Mo-Rh, and Rh-Rh, there is an increasing trend to use
other combinations such as WRh, WAg, and WAl with
digital mammography. These alternative W-target techniques
may allow for modest reduction in radiation dose with no
apparent reduction in image quality.5961 In addition, the exploration of techniques such as contrast-enhanced digital
mammography CEDM and DBT that may require x-ray
tubes with increased heat capacity and output also have an
influence on the choice of anode material. In addition, radiation dose reduction with photon counting detectors,62 a-Si:H
indirect conversion detectors,63,64 and CR65 have also been
reported. While phantom studies indicated the possibility of
50% dose reduction with digital mammography,66 studies
with clinical backgrounds indicate that such a drastic dose
reduction could adversely affect detection of microcalcifications and discrimination of masses substantially, and to a
lesser extent adversely affect detection of masses.67,68 Continuing technological improvements and technique refinements could lead to further dose reduction while maintaining
image quality.
Extensive investigations on radiation dose to the breast
and its dependence on breast composition, breast thickness,
and x-ray spectral characteristics have been documented.6972
While it was believed that radiation dose to other organs
including the uterus would be low during a mammography
exam, until recently quantitative estimates were
unavailable.73,74 A Monte Carlo computational study with an
anthropomorphic software phantom indicates that the dose to
the uterus during the first trimester, where a women may be
unaware of her pregnancy, is less than 0.03 Gy per imaged
breast during a bilateral two-view mammography exam.75
4881
III. ULTRASONOGRAPHY
The role of B-mode ultrasound in breast imaging has been
largely limited to applications such as distinguishing between cystic versus solid masses, evaluation of palpable
masses, and for needle core biopsy of masses. In recent
years, the number of indications has been greatly expanded
and breast ultrasonography is now an essential modality in
breast imaging. A center frequency above 10 MHz is now
recommended for adequate spatial resolution by the American College of Radiology ACR. B-mode imaging works
well for the established indications of breast ultrasonography,
Medical Physics, Vol. 35, No. 11, November 2008
4881
4882
4882
4883
4883
While there have been considerable advances in mammography, there is one major inherent limitation in that the
mammographic image reduces the three-dimensional
anatomy of the breast into a two-dimensional image. This
resulting superposition of normal breast structures, which
causes a visually distracting mask, is often referred to as
anatomical noise and has been shown to impair lesion
detection.192194 In addition, tissue superposition can mimic
the presence of a lesion, resulting in increased recall rates
and in increased biopsy rates, that add to the inconvenience
4884
4884
4885
4885
FIG. 2. A clinical case showing 1.6 cm low-grade invasive ductal carcinoma with minor ductal carcinoma in situ DCIS component. Better visualization of
the tumor is observed with DBT right, B compared to mammography left, A. Courtesy: Steven P. Poplack, Dartmouth Hitchcock Medical Center
4886
images, and indicated that CT imaging can significantly improve the confidence to detect mass but was inferior for
microcalcifications.231
In 2001, a landmark article by Boone et al. provided
quantitative estimates of radiation dose from a dedicated
BCT exam and image quality achieved with cadaveric
breast.232 The development of high-frame-rate flat-panel detectors accelerated further exploration of this technique.
Works addressing radiation dose to the imaged breast232234
and other organs,235 x-ray scatter,236,237 system design,238,239
imaging trajectory,240,241 image acquisition technique
factors,242244 resolution,245 noise,246 and artifacts 247 have
been published.
Independent studies with surgical mastectomy
specimens248 and with limited number of subjects249 suggest
that BCT provides for excellent anatomical detail and soft
tissue lesion visualization. Figure 3 shows multi-planar reconstructions acquired with a clinical prototype dedicated
BCT system and a MLO view projection image acquired
with a digital mammography system. Excellent 3-D visualization of the anatomy and soft tissue lesion is observed. A
recent study that compared visualization in BCT with lesions
detected in screen-film mammography in 65 subjects, suggest that BCT was superior for visualization of masses but
was inferior for visualization of microcalcifications.250
Some of the challenges with breast CT include simultaneous inclusion of medial and axillary aspects of the breast
during the scan and adequate visualization of
microcalcifications.250 Limitations in breast coverage with
dedicated breast CT has been inferential, and as yet there
have been no quantitative studies comparing tissue inclusion
with mammography. Improved breast coverage can be potentially achieved through system design including novel table
design251 and imaging trajectories,240,241 and with appropriate patient positioning. Current prototype systems used in
clinical studies utilize circular scan,250,252 and we are aware
of only one implementation using a non-circular trajectory
Medical Physics, Vol. 35, No. 11, November 2008
4886
While investigations into the applicability of digital subtraction angiography of the breast in the mid-1980s showed
the potential for differential diagnosis for malignant and benign tumors,260,261 the lack of appropriate imaging system
was a major limitation. The advent of digital mammography
has stimulated the exploration of quicker and cost-effective
alternative for breast MRI such as intravenously injected iodinated contrast media enhanced imaging of the breast for
angiogenesis imaging. Two approaches for contrastenhanced digital mammography are being explored. In dualenergy K-edge subtraction, two images at different energies
4887
X-ray phase sensitive imaging such as in-line phase contrast x-ray imaging278,279 and diffraction enhanced
imaging280283 are currently being investigated. The theoretical basis and design considerations for phase contrast
imaging284,285 and method to retrieve the phase map from
attenuation and phase contrast images286 have been well addressed. Development of a dual-detector system that uses
two CR cassettes for simultaneous acquisition of conventional attenuation and phase contrast images, from which
Medical Physics, Vol. 35, No. 11, November 2008
4887
4888
4888
FIG. 4. Conventional attenuation images A, C and phase contrast images B, D of accreditation phantom top row and lumpectomy specimen bottom
row. All images were acquired at 40 kVp with same entrance exposure for attenuation and phase contrast images. These images were acquired using
prototype systems developed at the University of Oklahoma Hong Liu, in collaboration with University of Alabama at Birmingham Xizeng Wu and
University of Iowa Laurie L. Fajardo. Courtesy: Hong Liu, University of Oklahoma
4889
4889
tively used with modalities that provide for anatomical structure such as x-ray, ultrasound, or MRI. At present, phasesensitive x-ray imaging that provides for improved contrast
and edge-enhancement has been demonstrated with specimens and would require substantial amount of translational
research to bring it to clinical use. From the technological
perspective, in addition to improvements with each modality,
we are likely to observe an increasing trend towards multimodality systems that combine the relative strengths of each
modality.
Importantly, we are likely to observe a paradigm shift in
the manner in which breast cancer screening will be performed in future. Breast cancer screening is currently performed in a standard manner for all women with mammography. However, this level of standardization is likely to be
replaced by a screening program where the selection of an
imaging modality or modalities would depend on an individuals risk and other classifications. Already elements of
this change can be observed with the recommendation by
American Cancer Society to adjunctively use breast MRI for
Medical Physics, Vol. 35, No. 11, November 2008
4890
4890
4891
54
76
4891
4892
97
P. C. Lauterbur, Image formation by induced local interactions: Examples employing nuclear magnetic resonance, Nature London 242,
190191 1973.
98
P. Mansfield, P. G. Morris, R. Ordidge, R. E. Coupland, H. M. Bishop,
and R. W. Blamey, Carcinoma of the breast imaged by nuclear magnetic
resonance NMR, Br. J. Radiol. 52615, 242243 1979.
99
R. J. Ross, J. S. Thompson, K. Kim, and R. A. Bailey, Nuclear magnetic
resonance imaging and evaluation of human breast tissue: preliminary
clinical trials, Radiology 1431, 195205 1982.
100
S. J. El Yousef, R. H. Duchesneau, R. J. Alfidi, J. R. Haaga, P. J. Bryan,
and J. P. LiPuma, Magnetic resonance imaging of the breast. Work in
progress, Radiology 1503, 761766 1984.
101
S. J. El Yousef, R. J. Alfidi, R. H. Duchesneau, C. A. Hubay, J. R. Haaga,
P. J. Bryan, J. P. LiPuma, and A. E. Ament, Initial experience with
nuclear magnetic resonance NMR imaging of the human breast, J.
Comput. Assist. Tomogr. 72, 215218 1983.
102
C. B. Stelling, P. C. Wang, A. Lieber, S. S. Mattingly, W. O. Griffen, and
D. E. Powell, Prototype coil for magnetic resonance imaging of the
female breast. Work in progress, Radiology 1542, 457462 1985.
103
R. C. Brasch, H. J. Weinmann, and G. E. Wesbey, Contrast-enhanced
NMR imaging: animal studies using gadolinium-DTPA complex, AJR,
Am. J. Roentgenol. 1423, 625630 1984.
104
D. H. Carr, J. Brown, G. M. Bydder, R. E. Steiner, H. J. Weinmann, U.
Speck, A. S. Hall, and I. R. Young, Gadolinium-DTPA as a contrast
agent in MRI: Initial clinical experience in 20 patients, AJR, Am. J.
Roentgenol. 1432, 215224 1984.
105
D. H. Carr, J. Brown, G. M. Bydder, H. J. Weinmann, U. Speck, D. J.
Thomas, and I. R. Young, Intravenous chelated gadolinium as a contrast
agent in NMR imaging of cerebral tumours, Lancet 18375, 484486
1984.
106
M. Laniado, H. J. Weinmann, W. Schorner, R. Felix, and U. Speck, First
use of GdDTPA/dimeglumine in man, Physiol. Chem. Phys. Med. NMR
162, 157165 1984.
107
H. J. Weinmann, R. C. Brasch, W. R. Press, and G. E. Wesbey, Characteristics of gadolinium-DTPA complex: a potential NMR contrast agent,
AJR, Am. J. Roentgenol. 1423, 619624 1984.
108
H. J. Weinmann, M. Laniado, and W. Mutzel, Pharmacokinetics of
GdDTPA/dimeglumine after intravenous injection into healthy volunteers, Physiol. Chem. Phys. Med. NMR 162, 167172 1984.
109
S. H. Heywang, D. Hahn, H. Schmidt, I. Krischke, W. Eiermann, R.
Bassermann, and J. Lissner, MR imaging of the breast using gadoliniumDTPA, J. Comput. Assist. Tomogr. 102, 199204 1986.
110
W. A. Kaiser and E. Zeitler, MR imaging of the breast: fast imaging
sequences with and without Gd-DTPA. Preliminary observations, Radiology 1703 Pt 1, 681686 1989.
111
J. Frahm, A. Haase, and D. Matthaei, Rapid NMR imaging of dynamic
processes using the FLASH technique, Magn. Reson. Med. 32, 321
327 1986.
112
J. Frahm, A. Haase, and D. Matthaei, Rapid three-dimensional MR imaging using the FLASH technique, J. Comput. Assist. Tomogr. 102,
363368 1986.
113
A. Haase, D. Matthaei, W. Hanicke, and J. Frahm, Dynamic digital subtraction imaging using fast low-angle shot MR movie sequence, Radiology 1602, 537541 1986.
114
J. Folkman and M. Klagsbrun, Angiogenic factors, Science 2354787,
442447 1987.
115
F. L. Flanagan, J. G. Murray, P. Gilligan, J. P. Stack, and J. T. Ennis,
Digital subtraction in Gd-DTPA enhanced imaging of the breast, Clin.
Radiol. 5012, 848854 1995.
116
C. D. Lehman, C. Gatsonis, C. K. Kuhl, R. E. Hendrick, E. D. Pisano, L.
Hanna, S. Peacock, S. F. Smazal, D. D. Maki, T. B. Julian, E. R. DePeri,
D. A. Bluemke, and M. D. Schnall, MRI evaluation of the contralateral
breast in women with recently diagnosed breast cancer, N. Engl. J. Med.
35613, 12951303 2007.
117
L. Liberman, E. A. Morris, C. M. Kim, J. B. Kaplan, A. F. Abramson, J.
H. Menell, K. J. Van Zee, and D. D. Dershaw, MR imaging findings in
the contralateral breast of women with recently diagnosed breast cancer,
AJR, Am. J. Roentgenol. 1802, 333341 2003.
118
S. G. Lee, S. G. Orel, I. J. Woo, E. Cruz-Jove, M. E. Putt, L. J. Solin, B.
J. Czerniecki, and M. D. Schnall, MR imaging screening of the contralateral breast in patients with newly diagnosed breast cancer: Preliminary results, Radiology 2263, 773778 2003.
119
C. D. Lehman, J. D. Blume, D. Thickman, D. A. Bluemke, E. Pisano, C.
Medical Physics, Vol. 35, No. 11, November 2008
4892
4893
152
4893
4894
172
4894
4895
231
4895
4896
4896
4897
guided diffuse optical spectroscopy of malignant and benign breast lesions, Neoplasia 44, 347354 2002.
295
B. Brooksby, S. Jiang, H. Dehghani, B. W. Pogue, K. D. Paulsen, J. B.
Weaver, C. Kogel, and S. P. Poplack, Combining near-infrared tomography and magnetic resonance imaging to study in vivo breast tissue:
Implementation of a Laplacian-type regularization to incorporate magnetic resonance structure, J. Biomed. Opt. 105, 051504 2005.
296
B. Brooksby, B. W. Pogue, S. Jiang, H. Dehghani, S. Srinivasan, C.
Kogel, T. D. Tosteson, J. Weaver, S. P. Poplack, and K. D. Paulsen,
Imaging breast adipose and fibroglandular tissue molecular signatures by
using hybrid MRI-guided near-infrared spectral tomography, Proc. Natl.
Acad. Sci. U.S.A. 10323, 88288833 2006.
297
C. M. Carpenter, B. W. Pogue, S. Jiang, H. Dehghani, X. Wang, K. D.
Paulsen, W. A. Wells, J. Forero, C. Kogel, J. B. Weaver, S. P. Poplack,
and P. A. Kaufman, Image-guided optical spectroscopy provides
molecular-specific information in vivo: MRI-guided spectroscopy of
breast cancer hemoglobin, water, and scatterer size, Opt. Lett. 328,
933935 2007.
298
V. Ntziachristos, A. G. Yodh, M. Schnall, and B. Chance, Concurrent
MRI and diffuse optical tomography of breast after indocyanine green
enhancement, Proc. Natl. Acad. Sci. U.S.A. 976, 27672772 2000.
299
X. Gu, Q. Zhang, M. Bartlett, L. Schutz, L. L. Fajardo, and H. Jiang,
Differentiation of cysts from solid tumors in the breast with diffuse
optical tomography, Acad. Radiol. 111, 5360 2004.
300
B. J. Tromberg, A. Cerussi, N. Shah, M. Compton, A. Durkin, D. Hsiang,
J. Butler, and R. Mehta, Imaging in breast cancer: diffuse optics in breast
cancer: detecting tumors in pre-menopausal women and monitoring neoadjuvant chemotherapy, Breast Cancer Research 76, 279285 2005.
301
N. Shah, J. Gibbs, D. Wolverton, A. Cerussi, N. Hylton, and B. J. Tromberg, Combined diffuse optical spectroscopy and contrast-enhanced
magnetic resonance imaging for monitoring breast cancer neoadjuvant
chemotherapy: a case study, J. Biomed. Opt. 105, 051503 2005.
302
D. B. Jakubowski, A. E. Cerussi, F. Bevilacqua, N. Shah, D. Hsiang, J.
Butler, and B. J. Tromberg, Monitoring neoadjuvant chemotherapy in
breast cancer using quantitative diffuse optical spectroscopy: A case
study, J. Biomed. Opt. 91, 230238 2004.
303
G. Boverman, Q. Fang, S. A. Carp, E. L. Miller, D. H. Brooks, J. Selb, R.
H. Moore, D. B. Kopans, and D. A. Boas, Spatio-temporal imaging of
the hemoglobin in the compressed breast with diffuse optical tomogra-
4897
Received 27 January 2008; revised 3 June 2008; accepted for publication 26 August 2008;
published 14 October 2008
PET/CT is an effective tool for the diagnosis, staging and restaging of cancer patients. It combines
the complementary information of functional PET images and anatomical CT images in one imaging session. Conventional stand-alone PET has been replaced by PET/CT for improved patient
comfort, patient throughput, and most importantly the proven clinical outcome of PET/CT over that
of PET and that of separate PET and CT. There are over two thousand PET/CT scanners installed
worldwide since 2001. Oncology is the main application for PET/CT. Fluorine-18 deoxyglucose is
the choice of radiopharmaceutical in PET for imaging the glucose uptake in tissues, correlated with
an increased rate of glycolysis in many tumor cells. New molecular targeted agents are being
developed to improve the accuracy of targeting different disease states and assessing therapeutic
response. Over 50% of cancer patients receive radiation therapy RT in the course of their disease
treatment. Clinical data have demonstrated that the information provided by PET/CT often changes
patient management of the patient and/or modifies the RT plan from conventional CT simulation.
The application of PET/CT in RT is growing and will become increasingly important. Continuing
improvement of PET/CT instrumentation will also make it easier for radiation oncologists to integrate PET/CT in RT. The purpose of this article is to provide a review of the current PET/CT
technology, to project the future development of PET and CT for PET/CT, and to discuss some
issues in adopting PET/CT in RT and potential improvements in PET/CT simulation of the thorax
in radiation therapy. 2008 American Association of Physicists in Medicine.
DOI: 10.1118/1.2986145
Key words: PET/CT, 4D CT, cine CT, MIP CT, average CT
I. INTRODUCTION
PET/CT was developed in 1998.1 It was not until 2001 when
the first commercial PET/CT scanner became available.2
Prior to the technology of PET/CT, the CT and the PET data
were acquired in two different scanners. Fusion of the PET
and CT data was performed with software techniques.3 Registration of the PET and CT data was more successful for the
brain studies with rigid transformation4 but less accurate for
the other regions of the body, in particular in the thorax and
the abdomen, due to the difficulty in repositioning the patient
in two separate sessions and the nonrigid nature of the
organs.1,5 Average error in the fusion of separate PET and CT
brain images was in the order of 2 3 mm.3 This error increased to 5 11 mm when fusing separate PET and CT body
images of the thorax.6 The advent of PET/CT scanners has
facilitated the hardware fusion of PET and CT data sets by
transporting the patient between the PET and CT components of the scanner without repeating patient setup. It has
been shown that hardware fusion is more accurate than software fusion in diagnosis, staging and restaging of many cancer types, in particular in the area of tumor infiltration of
adjacent structures that could not be conclusively assessed
using the separate CT and PET data.7
The fast scan speed of CT has shortened a normal imaging session of about 1 h with a stand-alone PET to less than
30 min with a PET/CT. This time savings has a major impact
on patient comfort in particular for patients who may need to
4955
raise their arms over the head during the PET/CT scan. The
CT images are adapted for attenuation correction and tumor
localization of the PET data.1 There are over 2000 PET/CT
scanners installed worldwide, and stand-alone PET scanners
have not been in production since 2006.5 Fluorine-18 deoxyglucose 18F-FDG is the major radiopharmaceutical in
PET/CT and has been approved for diagnosis and staging of
nonsmall-cell lung cancer NSCLC, colorectal cancer,
esophageal cancer, head and neck cancer, lymphoma, and
melanoma since 1998.8 In addition, PET/CT has been approved for staging and restaging and for therapeutic monitoring of breast cancer.
The average 18F-FDG PET sensitivity and specificity
across all oncology application are estimated at 84% based
on 18 402 patient studies and 88% based on 14 264 patient
studies, respectively, according to Gambhir et al.9 from a
collection of 419 articles from 1993 to 2000. Specifically, the
sensitivity of PET ranged from 84% to 86%, the specificity
ranged from 88% to 93%, and the accuracy ranged from 87%
to 90%.9 The addition of PET significantly improves the diagnosis of lung cancer with the sensitivity of 96% and specificity of 73% as compared to sensitivity of 67% for CT
alone, and the staging with the sensitivity of 83% and specificity of 91%, respectively as compared to sensitivity of
64% and specificity of 74% for CT alone.
The additional clinical values of PET/CT to PET alone or
separate PET and CT have also been documented. The first
0094-2405/2008/3511/4955/12/$23.00
4955
4956
4956
study was published by Kluetz et al.10 using the first prototype PET/CT scanner. It was found that hardware registration
enables physicians to more precisely discriminate between
physiologic and malignant 18F-FDG uptake and more accurately localize lesions. Hany et al.11 compared the data of
PET/CT and PET alone in 53 patients for the diagnosis or
suspicion of malignancy. Of 287 lesions, the sensitivity,
specificity, and accuracy increased from 90%, 93%, and 91%
for PET alone, to 98%, 99%, and 98% for PET/CT. BarShalom et al.12 evaluated 204 patients with 586 suspicious
lesions by interpreting PET and CT data together or PET and
CT data separately. They found that interpretation of PET
and CT together provided additional information over the
separate interpretation of PET and CT in 99 patients 49%
with 178 lesions 30%. PET/CT improved characterization
of equivocal lesions as definitely benign in 10% of sites and
as definitely malignant in 5% of sites. The results of PET/CT
had an impact on the management of 28 patients 14%.
Lardinois et al.13 compared the diagnostic accuracy of
PET/CT with that of CT alone, that of PET alone, and that of
conventional visual correlation of PET and CT in determining the stage of disease in 50 patients with proven or suspected NSCLC. PET/CT provided additional information in
20 of 49 patients. PET/CT had better diagnostic accuracy and
more accurate nodal staging than CT alone, PET alone, or
visual correlation of PET and CT.
Over 50% of all patients with cancer receive radiation
therapy RT during the course of their disease
management.14 Applications of PET to RT have been reported in head and neck,1526 lung,19,2740 breast,41
esophageal,42,43
non-Hodgkins
lymphoma,30,44
19,45
46,47
19
gynecologic,
rectal,
anal,
and prostate with
18
F-choline.48 Studies have also found that PET has advantages over CT in the standardization of volume
delineation,29,49,50 in the reduction of the risk for geometrical
misses,36 in the reduction of mediastinal nodal failure rate,51
in the minimization of radiation dose to the nontarget
organs,19,26,52 and in the assessment of tumor burden, blood
flow, tissue inflammation, and hypoxia.5358 Integration of
functional PET data with anatomical CT data should be a
standard in RT.59
There are some challenges in PET/CT imaging for RT.
The most challenging one is with reimbursement. Many patients are referred to a PET scan for diagnosis before receiving RT. The time between diagnosis and RT is too short for a
second PET to be reimbursable. In this situation, the diagnostic images of PET or PET/CT are fused using software
with the simulation CT images for RT. As of today, there is
no reimbursement for a simulation PET/CT. Unlike a whole
body PET/CT for diagnosis, a simulation PET/CT only needs
to be performed with a limited coverage much like in a simulation CT. A simulation PET/CT may be performed or embedded in a whole body PET/CT scanner for staging, restaging, or assessment of the treatment response in the course of
treatment. Integration of PET/CT in RT also involves at least
the following two disciplines of people working together:
Nuclear medicine and radiation oncology. Patient throughput
is an important concern in the practice of nuclear medicine,
4957
4957
Scintillator
Decay time ns
BGO
GSO
LSO, LYSO
15
25
80
300
60
40
2.4
3.3
2.7
4958
4958
4959
4959
FIG. 1. Effect of a patient motion at the 9th min during a single bed acquisition of 15 min. The CT before PET scan and PET images of the first 9 min were
in a and b, respectively. The CT after PET scan and PET images of the last 6 min were in c and d, respectively. The mean SUVs of the region of
interest pointed by the arrow were 2.7 in b, 1.2 in d and 2.1 in e of total 15 min acquisition.
4960
4960
(A)
(A)
(B)
(B)
(C)
FIG. 3. The PET/CT images of a 69 year-old female patient with an esophageal tumor after an induction chemotherapy. a shows an axial slice of the
fused clinical CT and PET image at the level of the esophageal tumor left
and the PET image in coronal view right. The radiology report indicated
the patient had a positive response to the chemotherapy. After removal of
misalignment by the AVG CT, the tumor reappeared in the same PET data
set in b. The arrows point to the tumor location. The gross target volumes
drawn in the images of a and b are shown in blue and in green, respectively, in c. The patient was treated with the tumor volume in green, and
the radiology report was corrected by the AVG CT.
4961
4961
FIG. 4. The RACT and the SSCT images of a patient with an average breath cycle of 4 s. The SSCT images in a were taken with one single CT gantry
rotation of 4 s, and the two images were 2.5 mm apart and 2.5 mm thick. The corresponding RACT images in b, obtained by averaging the cine CT images,
were averaged from 4 s of data collection over eight gantry rotations. The ACT images were almost free of reconstruction artifacts, which were observed in
the SSCT images reproduced from Fig. 11 of Ref. 79.
XI. 4D CT IMAGING
4D CT Refs. 122124 has found its acceptance in RT for
providing the gated CT images of multiple phases over a
respiratory cycle to assist contouring the extent of tumor
motion. 4D CT takes less time in acquisition than 4D PET
does. It normally takes less than 2 min like ACT to cover
40 cm in the superior-inferior direction on an 8- or 16-slice
CT. Two data acquisition modes have been used in 4D CT.
One is cine CT and the other one is low-pitch helical CT.
Both acquisitions scan any point in space for over one breath
cycle plus one or 2 / 3 gantry rotation cycle. Cine CT uses
less radiation and generates thinner slices than low-pitch helical CT. On the other hand, low-pitch helical CT has an
advantage over cine CT in acquisition time.131 The cine CT
based 4D CT is available on 4, 8, 16, and 64-slice CT scanners, and the low pitch helical CT based 4D CT is only
available on newer 16- and 64-slice CT scanners. Multislice
CT has significantly improved the speed of 4D CT data acquisition and made 4D CT clinically acceptable. It is expected that 4D CT will become an integral part of PET/CT
imaging of the thorax and 4D CT will be incorporated in the
quantification of 4D PET as each phase of gated PET image
needs its own gated CT image for attenuation
correction.108,132134 The ACT and MIP images can be derived from 4D CT or directly from cine CT.125
XII. DETERMINATION OF TUMOR CONTOUR IN
PET
Determination of tumor volume in PET image is normally
performed with SUV thresholds. This is an important yet
4962
4962
FIG. 5. The MIP CT and PET images of a patient. The MIP CT in a is displayed with window, level = 400, 40 and PET in b with the highest
intensity= 40% of the maximum SUV in the tumor. The images of a and b are displayed again with 1000, 700 in c and the highest density= 20% in
d, respectively. The tumor contour in c is superimposed in a, b, and d to demonstrate the effect on tumor size by display parameters.
controversial issue, in particular for the lung tumors impacted by the respiratory motion. To determine the gross tumor volume in PET, it is generally based on the visual interpretation by the experienced nuclear medicine physician.35
Some suggested an absolute threshold of 2.5,37,135 or a
threshold between 15% and 50% of the maximum SUV
mSUV.27,35,37,136138 Recent studies by Biehl et al.130 and
Nestle et al.78 suggested that no single SUV can reliably be
used for segmentation of the PET tumor volume, and that an
individualized threshold should be derived in consideration
of the size, location, nonuniform distribution of 18F-FDG
activity of the tumor. Most of the discussions have been with
the data of stand-alone PET. The issue may be further complicated in PET/CT. Any misalignment can further compromise the quantification of the PET data. Erdi et al.132 reported that different phases of 4D CT image can cause up to
24% variation of mSUV in the PET data. Pan et al.80 observed a variation of more than 50% mSUV between the
PET data with ACT and the same PET data with helical CT.
Chi et al.90 investigated the effects of misalignment to the
determination of PET GTV and found that GTV centroid
location could shift by 2.4 mm and GTV volume change
could be as high as 154% for the tumors of less than 50 cm3.
Partial volume also impacts the determination of SUV.
Partial volume is caused by the limited spatial resolution of
PET scanners 5 8 mm, and renders a blurry look of the
PET image. Partial volume mostly affects smaller tumor
sizes of less than 1 1.6 cm or 2 times the PET resolution. In
addition, studies have shown that SUV is a function of uptake time between injection of 18F-FDG and data
acquisition.61,139,140 Care should be taken to ensure the same
uptake time for the same patient imaged in a multiple of
PET/CT sessions during the course of diagnosis, treatment,
and staging. Tumor volume determined by PET may be more
indicative of the GTV plus motion since PET data are acquired over several min. PET GTV delineation has been and
Medical Physics, Vol. 35, No. 11, November 2008
4963
4963
4964
4964
planning for non-small cell lung cancer NSCLC, Lung Cancer 57,
125134 2007.
34
G. L. Ceresoli, G. M. Cattaneo, P. Castellone, G. Rizzos, C. Landoni, V.
Gregorc, R. Calandrino, E. Villa, C. Messa, A. Santoro, and F. Fazio,
Role of computed tomography and 18F fluorodeoxyglucose positron
emission tomography image fusion in conformal radiotherapy of nonsmall cell lung cancer: A comparison with standard techniques with and
without elective nodal irradiation, Tubercle 93, 8896 2007.
35
U. Nestle et al., 18F-deoxyglucose positron emission tomography FDGPET for the planning of radiotherapy in lung cancer: High impact in
patients with atelectasis, Int. J. Radiat. Oncol., Biol., Phys. 44, 593597
1999.
36
Y. E. Erdi, K. Rosenzweig, A. K. Erdi, H. A. Macapinlac, Y. C. Hu, L. E.
Braban, J. L. Humm, O. D. Squire, C. S. Chui, S. M. Larson, and E. D.
Yorke, Radiotherapy treatment planning for patients with non-small cell
lung cancer using positron emission tomography PET, Radiother. Oncol. 62, 5160 2002.
37
J. Bradley, W. L. Thorstad, S. Mutic, T. R. Miller, F. Dehdashti, B. A.
Siegel, W. Bosch, and R. J. Bertrand, Impact of FDG-PET on radiation
therapy volume delineation in non-small-cell lung cancer, Int. J. Radiat.
Oncol., Biol., Phys. 59, 7886 2004.
38
F. M. Kong, K. A. Frey, L. E. Quint, R. K. Ten Haken, J. A. Hayman, M.
Kessler, I. J. Chetty, D. Normolle, A. Eisbruch, and T. S. Lawrence, A
pilot study of 18Ffluorodeoxyglucose positron emission tomography
scans during and after radiation-based therapy in patients with non smallcell lung cancer, J. Clin. Oncol. 25, 31163123 2007.
39
M. P. Mac Manus, R. J. Hicks, D. L. Ball, V. Kalff, J. P. Matthews, E.
Salminen, P. Khaw, A. Wirth, D. Rischin, and A. McKenzie, F-18 fluorodeoxyglucose positron emission tomography staging in radical radiotherapy candidates with nonsmall cell lung carcinoma: Powerful correlation with survival and high impact on treatment, Cancer 92, 886895
2001.
40
M. T. Munley, L. B. Marks, C. Scarfone, G. S. Sibley, E. F. Patz, Jr., T. G.
Turkington, R. J. Jaszczak, D. R. Gilland, M. S. Anscher, and R. E.
Coleman, Multimodality nuclear medicine imaging in three-dimensional
radiation treatment planning for lung cancer: Challenges and prospects,
Lung Cancer 23, 105114 1999.
41
E. C. Ford, W. C. Lavely, D. A. Frassica, L. T. Myers, F. Asrari, R. L.
Wahl, and R. C. Zellars, Comparison of FDG-PET/CT and CT for delineation of lumpectomy cavity for partial breast irradiation, Int. J. Radiat. Oncol., Biol., Phys. 71, 595602 2008.
42
C. Everitt and T. Leong, Influence of F-fluorodeoxyglucose-positron
emission tomography on computed tomography-based radiation treatment
planning for oesophageal cancer, Australas Radiol. 50, 271274 2006.
43
T. Leong et al., A prospective study to evaluate the impact of FDG-PET
on CT-based radiotherapy treatment planning for oesophageal cancer,
Radiother. Oncol. 78, 254261 2006.
44
M. Hutchings, A. Loft, M. Hansen, A. K. Berthelsen, and L. Specht,
Clinical impact of FDG-PET/CT in the planning of radiotherapy for
early-stage Hodgkin lymphoma, Eur. J. Haematol. 78, 206212 2007.
45
J. Esthappan, S. Chaudhari, L. Santanam, S. Mutic, J. Olsen, D. M. Macdonald, D. A. Low, A. K. Singh, and P. W. Grigsby, Prospective clinical
trial of positron emission tomography/computed tomography imageguided intensity-modulated radiation therapy for cervical carcinoma with
positive para-aortic lymph nodes, Int. J. Radiat. Oncol., Biol., Phys. in
press.
46
I. F. Ciernik, M. Huser, C. Burger, J. B. Davis, and G. Szekely, Automated functional image-guided radiation treatment planning for rectal
cancer, Int. J. Radiat. Oncol., Biol., Phys. 62, 893900 2005.
47
D. A. Patel, S. T. Chang, K. A. Goodman, A. Quon, B. Thorndyke, S. S.
Gambhir, A. McMillan, B. W. Loo, Jr., and A. C. Koong, Impact of
integrated PET/CT on variability of target volume delineation in rectal
cancer, Technol. Cancer Res. Treat. 6, 3136 2007.
48
I. F. Ciernik, D. W. Brown, D. Schmid, T. Hany, P. Egli, and J. B. Davis,
3D-segmentation of the 18F-choline PET signal for target volume definition in radiation therapy of the prostate, Technol. Cancer Res. Treat. 6,
2330 2007.
49
R. J. Steenbakkers et al., Reduction of observer variation using matched
CT-PET for lung cancer delineation: A three-dimensional analysis, Int. J.
Radiat. Oncol., Biol., Phys. 64, 435448 2006.
50
J. L. Fox, R. Rengan, W. OMeara, E. Yorke, Y. Erdi, S. Nehmeh, S. A.
Leibel, and K. E. Rosenzweig, Does registration of PET and planning
CT images decrease interobserver and intraobserver variation in delineat-
ing tumor volumes for non-small-cell lung cancer?, Int. J. Radiat. Oncol., Biol., Phys. 62, 7075 2005.
51
D. De Ruysscher et al., Selective mediastinal node irradiation based on
FDG-PET scan data in patients with non-small-cell lung cancer: A prospective clinical study, Int. J. Radiat. Oncol., Biol., Phys. 62, 988994
2005.
52
A. van Baardwijk, B. G. Baumert, G. Bosmans, M. van Kroonenburgh, S.
Stroobants, V. Gregoire, P. Lambin, and D. De Ruysscher, The current
status of FDG-PET in tumour volume definition in radiotherapy treatment
planning, Cancer Treat Res. 32, 245260 2006.
53
R. D. Stewart and X. A. Li, BGRT: Biologically guided radiation
therapyThe future is fast approaching, Med. Phys. 34, 37393751
2007.
54
C. C. Ling, J. Humm, S. Larson, H. Amols, Z. Fuks, S. Leibel, and J. A.
Koutcher, Towards multidimensional radiotherapy MD-CRT: Biological imaging and biological conformality, Int. J. Radiat. Oncol., Biol.,
Phys. 47, 551560 2000.
55
C. C. Ling and X. A. Li, Over the next decade the success of radiation
treatment planning will be judged by the immediate biological response
of tumor cells rather than by surrogate measures such as dose maximization and uniformity, Med. Phys. 32, 21892192 2005.
56
K. S. Chao, W. R. Bosch, S. Mutic, J. S. Lewis, F. Dehdashti, M. A.
Mintun, J. F. Dempsey, C. A. Perez, J. A. Purdy, and M. J. Welch, A
novel approach to overcome hypoxic tumor resistance: Cu-ATSM-guided
intensity-modulated radiation therapy, Int. J. Radiat. Oncol., Biol., Phys.
49, 11711182 2001.
57
Z. Lin, J. Mechalakos, S. Nehmeh, H. Schoder, N. Lee, J. Humm, and C.
C. Ling, The influence of changes in tumor hypoxia on dose-painting
treatment plans based on 18F-FMISO positron emission tomography,
Int. J. Radiat. Oncol., Biol., Phys. 70, 12191228 2008.
58
I. Serganova, J. Humm, C. Ling, and R. Blasberg, Tumor hypoxia imaging, Clin. Cancer Res. 12, 52605264 2006.
59
K. Mah, C. B. Caldwell, Y. C. Ung, C. E. Danjoux, J. M. Balogh, S. N.
Ganguli, L. E. Ehrlich, and R. Tirona, The impact of 18FDG-PET on
target and critical organs in CT-based treatment planning of patients with
poorly defined non-small-cell lung carcinoma: A prospective study, Int.
J. Radiat. Oncol., Biol., Phys. 52, 339350 2002.
60
K. R. Zasadny and R. L. Wahl, Standardized uptake values of normal
tissues at PET with 2-fluorine-18-fluoro-2-deoxy-D-glucose: Variations
with body weight and a method for correction, Radiology 189, 847850
1993.
61
J. W. Keyes, Jr., SUV: Standard uptake or silly useless value?, J. Nucl.
Med. 36, 18361839 1995.
62
G. W. Goerres, C. Burger, M. R. Schwitter, T. N. Heidelberg, B. Seifert,
and G. K. von Schulthess, PET/CT of the abdomen: Optimizing the
patient breathing pattern, Eur. Radiol. 13, 734739 2003.
63
T. Beyer, G. Antoch, T. Blodgett, L. F. Freudenberg, T. Akhurst, and S.
Mueller, Dual-modality PET/CT imaging: The effect of respiratory motion on combined image quality in clinical oncology, Eur. J. Nucl. Med.
Mol. Imaging 30, 588596 2003.
64
G. W. Goerres, C. Burger, E. Kamel, B. Seifert, A. H. Kaim, A. Buck, T.
C. Buehler, and G. K. Von Schulthess, Respiration-induced attenuation
artifact at PET/CT: Technical considerations, Radiology 226, 906910
2003.
65
T. Beyer, S. Rosenbaum, P. Veit, J. Stattaus, S. P. Muller, F. P. Difilippo,
H. Schoder, O. Mawlawi, F. Roberts, A. Bockisch, and H. Kuhl, Respiration artifacts in whole-body 18F-FDG PET/CT studies with combined
PET/CT tomographs employing spiral CT technology with 1 to 16 detector rows, Eur. J. Nucl. Med. Mol. Imaging 32, 14291439 2005.
66
G. W. Goerres, T. F. Hany, E. Kamel, G. K. von Schulthess, and A. Buck,
Head and neck imaging with PET and PET/CT: Artefacts from dental
metallic implants, Eur. J. Nucl. Med. Mol. Imaging 29, 367370 2002.
67
G. W. Goerres, D. T. Schmid, and G. K. Eyrich, Do hardware artefacts
influence the performance of head and neck PET scans in patients with
oral cavity squamous cell cancer?, Dentomaxillofac Radiol. 32, 365371
2003.
68
E. M. Kamel, C. Burger, A. Buck, G. K. von Schulthess, and G. W.
Goerres, Impact of metallic dental implants on CT-based attenuation
correction in a combined PET/CT scanner, Eur. Radiol. 13, 724728
2003.
69
S. Bujenovic, F. Mannting, R. Chakrabarti, and D. Ladnier, Artifactual
2-deoxy-2-18Ffluoro-D-glucose localization surrounding metallic objects in a PET/CT scanner using CT-based attenuation correction, Mol.
4965
4965
correction of the PET data from PET/CT, Med. Phys. 34, 20392047
2007.
89
M. M. Osman, C. Cohade, Y. Nakamoto, L. T. Marshall, J. P. Leal, and R.
L. Wahl, Clinically significant inaccurate localization of lesions with
PET/CT: Frequency in 300 patients, J. Nucl. Med. 44, 240243 2003.
90
P. M. Chi, O. Mawlawi, D. Luo, Z. Liao, H. A. Macapinlac, and T. Pan,
Effects of respiration-averaged CT on PET/CT quantification and its
potential impact on GTV delineation, Int. J. Radiat. Oncol., Biol., Phys.
accepted.
91
D. W. Townsend, A combined PET/CT scanner: The choices, J. Nucl.
Med. 42, 533534 2001.
92
D. W. Townsend, Physical principles and technology of clinical PET
imaging, Ann. Acad. Med. Singapore 33, 133145 2004.
93
D. W. Townsend, T. Beyer, and T. M. Blodgett, PET/CT scanners: A
hardware approach to image fusion, Semin Nucl. Med. 33, 193204
2003.
94
D. W. Townsend, J. P. Carney, J. T. Yap, and N. C. Hall, PET/CT today
and tomorrow, J. Nucl. Med., Suppl. 45, 4S14S 2004.
95
D. W. Townsend and S. R. Cherry, Combining anatomy and function:
The path to true image fusion, Eur. Radiol. 11, 19681974 2001.
96
S. R. Cherry, The 2006 Henry N. Wagner lecture: Of mice and men and
positronsAdvances in PET imaging technology, J. Nucl. Med. 47,
17351745 2006.
97
T. M. Blodgett, C. C. Meltzer, and D. W. Townsend, PET/CT: Form and
function, Radiology 242, 360385 2007.
98
J. R. Stickel and S. R. Cherry, High-resolution PET detector design:
modelling components of intrinsic spatial resolution, Phys. Med. Biol.
50, 179195 2005.
99
O. Mawlawi, D. A. Podoloff, S. Kohlmyer, J. J. Williams, C. W. Stearns,
R. F. Culp, and H. Macapinlac, Performance characteristics of a newly
developed PET/CT scanner using NEMA standards in 2D and 3D
modes, J. Nucl. Med. 45, 17341742 2004.
100
S. Surti, A. Kuhn, M. E. Werner, A. E. Perkins, J. Kolthammer, and J. S.
Karp, Performance of Philips Gemini TF PET/CT scanner with special
consideration for its time-of-flight imaging capabilities, J. Nucl. Med.
48, 471480 2007.
101
P. E. Kinahan and J. S. Karp, Figures of merit for comparing reconstruction algorithms with a volume-imaging PET scanner, Phys. Med. Biol.
39, 631642 1994.
102
R. Leahy and C. Byrne, Recent developments in iterative image reconstruction for PET and SPECT, IEEE Trans. Med. Imaging 19, 257260
2000.
103
V. Bettinardi, P. Mancosu, M. Danna, G. Giovacchini, C. Landoni, M.
Picchio, M. C. Gilardi, A. Savi, I. Castiglioni, M. Lecchi, and F. Fazio,
Two-dimensional vs three-dimensional imaging in whole body oncologic PET/CT: A discovery-STE phantom and patient study, Q. J. Nucl.
Med. Mol. Imaging 51, 214223 2007.
104
E. J. Hoffman, M. E. Phelps, G. Wisenberg, H. R. Schelbert, and D. E.
Kuhl, Electrocardiographic gating in positron emission computed tomography, J. Comput. Assist. Tomogr. 3, 733739 1979.
105
S. A. Nehmeh, Y. E. Erdi, C. C. Ling, K. E. Rosenzweig, H. Schoder, S.
M. Larson, H. A. Macapinlac, O. D. Squire, and J. L. Humm, Effect of
respiratory gating on quantifying PET images of lung cancer, J. Nucl.
Med. 43, 876881 2002.
106
S. A. Nehmeh, Y. E. Erdi, C. C. Ling, K. E. Rosenzweig, O. D. Squire, L.
E. Braban, E. Ford, K. Sidhu, G. S. Mageras, S. M. Larson, and J. L.
Humm, Effect of respiratory gating on reducing lung motion artifacts in
PET imaging of lung cancer, Med. Phys. 29, 366371 2002.
107
S. A. Nehmeh, Y. E. Erdi, K. E. Rosenzweig, H. Schoder, S. M. Larson,
O. D. Squire, and J. L. Humm, Reduction of respiratory motion artifacts
in PET imaging of lung cancer by respiratory correlated dynamic PET:
Methodology and comparison with respiratory gated PET, J. Nucl. Med.
44, 16441648 2003.
108
S. A. Nehmeh et al., Four-dimensional 4D PET/CT imaging of the
thorax, Med. Phys. 31, 31793186 2004.
109
P. E. Kinahan, H. Vesselle, L. MacDonald, A. M. Alessio, S. Kohlmyer,
and T. Lewellen, Whole-body respiratory gated PET/CT, J. Nucl. Med.
Meeting Abstracts, May 2006; 47, 187P.
110
O. Mawlawi, T. Pan, and H. A. Macapinlac, PET/CT imaging techniques, considerations, and artifacts, J. Thorac. Imaging 21, 99110
2006.
111
J. L. Garcia-Ramirez, S. Mutic, J. F. Dempsey, D. A. Low, and J. A.
Purdy, Performance evaluation of an 85-cm-bore x-ray computed tomog-
70
4966
raphy scanner designed for radiation oncology and comparison with current diagnostic CT scanners, Int. J. Radiat. Oncol., Biol., Phys. 52, 1123
1131 2002.
112
T. Beyer, A. Bockisch, H. Kuhl, and M. J. Martinez, Whole-body 18FFDG PET/CT in the presence of truncation artifacts, J. Nucl. Med. 47,
9199 2006.
113
J. S. Maltz, S. Bose, H. P. Shukla, and A. R. Bani-Hashemi, CT truncation artifact removal using water-equivalent thicknesses derived from
truncated projection data, Conf. Proc. IEEE Eng. Med. Biol. Soc. 2007,
29072911 2007.
114
O. Mawlawi, J. J. Erasmus, T. Pan, D. D. Cody, R. Campbell, A. H. Lonn,
S. Kohlmyer, H. A. Macapinlac, and D. A. Podoloff, Truncation artifact
on PET/CT: Impact on measurements of activity concentration and assessment of a correction algorithm, AJR, Am. J. Roentgenol. 186, 1458
1467 2006.
115
K. R. Britton, G. Starkschall, S. L. Tucker, T. Pan, C. Nelson, J. Y. Chang,
J. D. Cox, R. Mohan, and R. Komaki, Assessment of gross tumor volume regression and motion changes during radiotherapy for non-smallcell lung cancer as measured by four-dimensional computed tomography, Int. J. Radiat. Oncol., Biol., Phys. in press.
116
W. V. Vogel, J. A. van Dalen, B. Wiering, H. Huisman, F. H. Corstens, T.
J. Ruers, and W. J. Oyen, Evaluation of image registration in PET/CT of
the liver and recommendations for optimized imaging, J. Nucl. Med. 48,
910919 2007.
117
M. M. Osman, C. Cohade, Y. Nakamoto, and R. L. Wahl, Respiratory
motion artifacts on PET emission images obtained using CT attenuation
correction on PET-CT, Eur. J. Nucl. Med. Mol. Imaging 30, 603606
2003.
118
G. W. Goerres, E. Kamel, B. Seifert, C. Burger, A. Buck, T. F. Hany, and
G. K. Von Schulthess, Accuracy of image coregistration of pulmonary
lesions in patients with non-small cell lung cancer using an integrated
PET/CT system, J. Nucl. Med. 43, 14691475 2002.
119
G. W. Goerres, E. Kamel, T. N. Heidelberg, M. R. Schwitter, C. Burger,
and G. K. von Schulthess, PET-CT image co-registration in the thorax:
influence of respiration, Eur. J. Nucl. Med. Mol. Imaging 29, 351360
2002.
120
C. B. Caldwell, K. Mah, M. Skinner, and C. E. Danjoux, Can PET
provide the 3D extent of tumor motion for individualized internal target
volumes? A phantom study of the limitations of CT and the promise of
PET, Int. J. Radiat. Oncol., Biol., Phys. 55, 13811393 2003.
121
P. J. Keall et al., The management of respiratory motion in radiation
oncology report of AAPM Task Group 76, Med. Phys. 33, 38743900
2006.
122
D. A. Low et al., A method for the reconstruction of four-dimensional
synchronized CT scans acquired during free breathing, Med. Phys. 30,
12541263 2003.
123
T. Pan, T. Y. Lee, E. Rietzel, and G. T. Chen, 4D-CT imaging of a
volume influenced by respiratory motion on multi-slice CT, Med. Phys.
31, 333340 2004.
124
P. J. Keall, G. Starkschall, H. Shukla, K. M. Forster, V. Ortiz, C. W.
Stevens, S. S. Vedam, R. George, T. Guerrero, and R. Mohan, Acquiring
4D thoracic CT scans using a multislice helical method, Phys. Med.
Biol. 49, 20532067 2004.
125
T. Pan, X. Sun, and D. Luo, Improvement of the cine-CT based 4D-CT
imaging, Med. Phys. 34, 44994503 2007.
4966
126
Christopher J. Amies
Siemens Medical Solutions USA, Inc., Oncology Care Systems Group, 4040 Nelson Avenue, Concord,
California 94520
Michelle Svatos
Translational Research, Varian Medical Systems, 3100 Hansen Way M/S E263 Palo Alto,
California 94304-1038
Received 9 May 2008; revised 11 August 2008; accepted for publication 18 September 2008;
published 6 November 2008
Intensity modulated radiation therapy IMRT is an advanced form of external beam radiation
therapy. IMRT offers an additional dimension of freedom as compared with field shaping in threedimensional conformal radiation therapy because the radiation intensities within a radiation field
can be varied according to the preferences of locations within a given beam direction from which
the radiation is directed to the tumor. This added freedom allows the treatment planning system to
better shape the radiation doses to conform to the target volume while sparing surrounding normal
structures. The resulting dosimetric advantage has shown to translate into clinical advantages of
improving local and regional tumor control. It also offers a valuable mechanism for dose escalation
to tumors while simultaneously reducing radiation toxicities to the surrounding normal tissue and
sensitive structures. In less than a decade, IMRT has become common practice in radiation oncology. Looking forward, the authors wonder if IMRT has matured to such a point that the room for
further improvement has diminished and so it is pertinent to ask what the future will hold for IMRT.
This article attempts to look from the perspective of the current state of the technology to predict
the immediate trends and the future directions. This article will 1 review the clinical experience of
IMRT; 2 review what we learned in IMRT planning; 3 review different treatment delivery
techniques; and finally, 4 predict the areas of advancements in the years to come. 2008 American Association of Physicists in Medicine. DOI: 10.1118/1.3002305
Key words: radiation therapy, intensity modulation, treatment planning, radiotherapy delivery,
IMRT
I. OVERVIEW
Intensity modulated radiation therapy IMRT has been
widely adopted as a new tool in radiation therapy to deliver
high doses of radiation to the tumor while providing maximal sparing of surrounding critical structures. What facilitated the quick dissemination of IMRT technology was not
hard clinical evidence but rather the individually calculated
and visually illustrated dosimetric advantages. Because such
dosimetric advantage was obvious, wide clinical adoption of
IMRT preceded any randomized clinical trials. In the U.S.,
the increased reimbursement for the technology also fueled
the speed of clinical dissemination. Early studies indicated
that with IMRT, radiation doses to sensitive structures could
be reduced significantly while maintaining sufficient dose
coverage to the targeted tumorous tissues.19 Both rotational
and gantry-fixed IMRT techniques have been implemented
clinically
using
dynamic
multileaf
collimation
DMLC.1,36,1014 In gantry-fixed IMRT, multiple coplanar
and noncoplanar beams at different orientations, each with
spatially modulated beam intensities, are used.1,3,12 Rotational IMRT, as it is practiced today, mainly employs temporally modulated fan beams,5,6 commonly known as tomo5233
0094-2405/2008/3512/5233/9/$23.00
5233
5234
5234
5235
the users clinical goals.78 The resultant plans from this approach are also called Pareto-optimal plans.
One of the tasks in treatment planning for external beam
radiation therapy is to determine the number of beams to use
and their orientations. For conventional treatments, beam
angles used for different treatment sites are well established.
With IMRT, the task of beam angle selection is more complicated and less intuitive because of more complex interactions and mutual compensations among the different beam
angles as the result of computer optimized beam intensities.
As the result, beam angle optimization has been one of the
areas of intensive investigation. Haas et al.79 employed genetic algorithms to search for the best beam orientations.
Rowbottom et al.80 and Stein et al.81 used simulated annealing algorithms to perform beam angle optimization by comparing thousands of sets of fixed number of beams sampled
from a constrained or unconstrained space of beam orientations. Pugachev et al.82 also reported different schemes of
beam angle optimization with the simulated annealing algorithm. Although all these studies demonstrated some effects
of beam angle selection on plan quality, the degree of improvements has been small. It is thus reasonable to conclude
that the mutual interactions and compensations among the
beams under intensity optimization also made beam angle
selection less influential on plan quality as compared with
three-dimensional conformal therapy.
In spite of years of refinements in IMRT planning, arguably, the quality of treatment plans has not improved much
from that of the early days. The explanation may be rooted in
the basic principles of inverse planning. For each patient, the
anatomy dictates a unique set of preferred beam orientations
and, in each beam orientation and radiation field, the preferred locations through which radiation is directed to the
tumor. If more freedom is given to make use of these intrinsic preferences in treatment planning, either performed by a
human planner or by a computer optimization system, better
plans can be generated. IMRT provides an additional freedom as compared with 3D conformal radiation therapy because the radiation intensities within a radiation field can be
varied according to the location preferences. However, there
is a fundamental limit to which the quality of treatment plans
can be improved. This limit is defined by the physics of the
radiation dose deposition and by the degrees of freedom that
are given to the computer optimization routine. There are
indications that the required degree of intensity variation for
achieving the optimal plan quality for a large majority of
cases is relatively small. For example, Ludlum et al.83 optimized IMRT plans for different challenging cancer sites using different number of intensity levels. They found that reducing the number of intensity levels from 10 to 3 only cause
minor degradations in dose distribution. Comparisons among
different treatment planning and delivery approaches8487
have not yield any approach with clinically meaningful superiority. These studies and our own experience led us to
believe that with the current state of the radiation delivery
systems and without new advances to provide additional degrees of freedom, IMRT has approached its limits in the
quality of treatment plans that can be physically achieved. As
Medical Physics, Vol. 35, No. 12, December 2008
5235
5236
5236
The fact that treatment plans of different complexity designed for different delivery methods can achieve similar
quality of treatments suggest that there is substantial room
for improving the workflow and efficiency. Past attempts in
improving treatment efficiency include the use of direct aperture optimization95 and the development of single arc
IMAT.96100
Instead of optimizing intensity maps and then translating
the intensity maps into deliverable segments, Shepard et al.95
proposed and developed a method called direct aperture optimization DAO that directly optimizes the shapes and
weights of MLC segments simultaneously in one step. The
planner determines the number of beams to use and the num-
5237
5237
Besides improving efficiency, great opportunities also exist for improving the accuracy of treatment delivered by using on-board image guidance. The advent of on-board cone
beam computed tomography CBCT and in-room CT with
high soft tissue contrast opens new opportunities for highprecision radiation therapy.106 The success of the image
guided radiation therapy IGRT lies not only in the ability to
acquire the images but largely in how we use the images to
achieve our goal of controlling more cancers while decreasing the normal tissue toxicities. In many cases, the tumor not
only changes its location but also its shape and the relative
geometrical relationship with its surrounding normal structures. Consequently, simply shifting the patient does not
guarantee optimal treatments. Therefore, reoptimizing the
IMRT plan or adapting an existing plan to the anatomy of
the day will increasingly become an active area of research.
Recognizing the impracticality of a full-fledged daily replanning with current technology, several researchers have
recently proposed methods for incorporating the deformable
component into an online correction strategy. Wu et al.107
have shown that for tomotherapy delivery, intensities can be
modified online with given transformation parameters or
with a quick reoptimization. Mohan et al.108 proposed to deform the intensity patterns as an online correction strategy.
Feng et al.109 have proposed a scheme that deforms the aperture shapes of the IMRT segments by using the deformation matrix derived from the planning image set and the images of the day. Ahunbay et al.110 developed a similar
scheme but added aperture weight optimization after aperture
deformation. These schemes have shown to be able to approach the plan quality resulting from reoptimization using
the images of the day.
As computer technology continue to advance, such short
cuts as described above may no longer be needed. Replanning using the images of the day will become a reality. The
advance of IGRT will make imaging, planning and treatment
delivery all performed in a single session. Under the new
image-plan-treat process, initial imaging and planning will
become merely a first look into what degree of dose conformity is achievable. With experience and confidence, the initial planning may even be eliminated. Such a procedure,
which has been the norm for radiosurgery, will allow further
shrinkage of PTV margins and encourage radiation oncologists to revise the current dose fractionation schemes. There
are three technologies that will enable for this image-plantreat scheme.
1 Imaging systems integrated with delivery machines will
be faster and provide greater image quality before and
during treatment. Currently, the gantry speed is setting
the limit on the speed of image acquisition for imaging
systems mounted on the treatment gantry. The image
quality achievable with CBCT also does not match that
of fan beam CT units because of the limitations of the
reconstruction algorithms for CBCT and the added radiation scatter contributions. These limitations will be
5238
corrected in the long-term by the emergence of new designs of on-line imaging systems, new imaging reconstruction and scatter subtraction algorithms, and/or new
on-line imaging strategies.
2 Dose calculation and optimization can be performed
within a couple of minutes. Computational speeds have
been increasing rapidly according to Moores law. Other
innovations, such as the use of special hardware for dose
computation,111 further push the envelope of planning
speed.
3 Fast image registration will allow the structures delineated by the physicians on a previously acquired image
set to be transferred to the images of the day in very
short time. Although three-dimensional deformable image registration has been theoretically solved for decades, practical applications of these algorithms in radiation therapy are new and relatively primitive. The room
of improvements in both the speed and the accuracy of
registration is significant. As the need for speed and accuracy continue to rise, innovative solutions will continue to emerge. It will be possible within the next
5 years for a deformable image registration to be performed in less than a minute. With fast deformable image registration, the accumulation of doses to different
parts of the target and to critical structures will be performed more accurately.
V. CONCLUSION
We have reviewed and described the current state of
IMRT and identified that IMRT is a well established technology offering dosimetric advantages for target coverage and
normal tissue avoidance. We believe that the treatment plan
quality has reached a limit imposed by the physics of photon
dose deposition and the room for further improvement in
plan quality is small. We also noticed that this intrinsic limit
is also not difficult to approach with different IMRT
schemes. This fact has allowed efficiency improvements
Medical Physics, Vol. 35, No. 12, December 2008
5238
5239
19
5239
40
J. C. Landry et al., Treatment of pancreatic cancer tumors with intensitymodulated radiation therapy IMRT using the volume at risk approach
VARA: Employing dose-volume histogram DVH and normal tissue
complication probability NTCP to evaluate small bowel toxicity, Med.
Dosim. 27, 121129 2002.
41
C. M. Nutting et al., A comparison of conformal and intensitymodulated techniques for esophageal radiotherapy, Radiother. Oncol. 61,
157163 2001.
42
R. B. Kuppersmith et al., The use of intensity modulated radiotherapy
for the treatment of extensive and recurrent juvenile angiofibroma, Int. J.
Pediatr. Otorhinolaryngol. 52, 261268 2000.
43
A. C. Paulino and M. Skwarchuk Intensity-modulated radiation therapy
in the treatment of children, Med. Dosim. 27, 115120 2002.
44
E. Huang et al., Intensity-modulated radiation therapy for pediatric
medulloblastoma: Early report on the reduction of ototoxicity, Int. J.
Radiat. Oncol., Biol., Phys. 52, 599605 2002.
45
L. Hong, K. M. Alektiar, M. Hunt, E. Venkatraman, and S. A. Leibel,
Intensity-modulated radiotherapy for soft tissue sarcoma of the thigh,
Int. J. Radiat. Oncol., Biol., Phys. 593,752975 2004.
46
D. M. Shepard, G. Olivera, L. Angelos, O. Sauer, P. Reckwerdt, and T. R.
Mackie, A simple model for examining issues in radiotherapy optimization, Med. Phys. 267, 12121221 1999.
47
E. J. Hall and C. Wuu, Radiation-induced second cancers: The impact of
3D-CRT and IMRT, Int. J. Radiat. Oncol., Biol., Phys. 56, 8388 2003.
48
D. B. Mansur, E. E. Klein, and B. P. Maserang, Measured peripheral
dose in pediatric radiation therapy: A comparison of intensity-modulated
and conformal techniques, Radiother. Oncol. 822, 179184 2007.
49
A. M. Allen et al., Fatal pneumonitis associated with intensitymodulated radiation therapy for mesothelioma, Int. J. Radiat. Oncol.,
Biol., Phys. 65, 640645 2006.
50
D. C. Rice, W. R. Smythe, Z. Liao, T. Guerrero, J. Y. Chang, M. F.
McAleer, M. D. Jeter, A. Correa, A. A. Vaporciyan, H. H. Liu, R. Komaki, K. M. Forster, and C. W. Stevens, Dose-dependent pulmonary
toxicity after postoperative intensity-modulated radiotherapy for malignant pleural mesothelioma, Int. J. Radiat. Oncol., Biol., Phys. 692,
350357 2007.
51
C. X. Yu, D. A. Jaffray, and J. W. Wong, Effects of intra-fraction organ
motion on the delivery of dynamic intensity modulation, Phys. Med.
Biol. 43, 91104 1998.
52
H. D. Kubo and B. C. Hill, Respiration-gated radiotherapy treatment: A
technical study, Phys. Med. Biol. 41, 8391 1996.
53
P. J. Keall, V. R. Kini, S. S. Vedam, and R. Mohan, Motion adaptive
x-ray therapy: A feasibility study, Phys. Med. Biol. 46, 110 2001.
54
W. D. DSouza, S. A. Naqvi, and C. X. Yu, Real-time intra-fraction
motion tracking using the treatment couch: A feasibility study, Phys.
Med. Biol. 50, 40214033 2005.
55
D. A. Kuban, S. L. Tucker, L. Dong, G. Starkschall, E. H. Huang, M. R.
Cheung, A. K. Lee, and A. Pollack, Long-term results of the M. D.
Anderson randomized dose-escalation trial for prostate cancer, Int. J.
Radiat. Oncol., Biol., Phys. 701, 6774 2008.
56
A. Pollack, A. Hanlon, E. M. Horwitz, S. Feigenberg, R. G. Uzzo, and R.
A. Price, Radiation therapy dose escalation for prostate cancer: A rationale for IMRT, World J. Urol. 4, 200208 2003.
57
M. J. Zelefsky, Z. Fuks, and S. A. Leibel, Intensity-modulated radiation
therapy for prostate cancer, Semin. Radiat. Oncol. 3, 229237 2002.
58
S. Junius, K. Haustermans, B. Bussels, R. Oyen, B. Vanstraelen, T.
Depuydt, J. Verstraete, S. Joniau, and H. Van Poppel, Hypofractionated
intensity modulated irradiation for localized prostate cancer, results from
a phase I/II feasibility study, Radiother. Oncol. 2, 29 2007.
59
K. K. Fu et al., A Radiation Therapy Oncology Group RTOG phase III
randomized study to compare hyperfractionation and two variants of accelerated fractionation to standard fractionation radiotherapy for head and
neck squamous cell carcinomas: First report of RTOG 9003, Int. J. Radiat. Oncol., Biol., Phys. 48, 716 2000.
60
G. M. Freedman, N. J. Meropol, E. R. Sigurdson, J. Hoffman, E. Callahan, R. Price, J. Cheng, S. Cohen, N. Lewis, D. Watkins-Bruner, A.
Rogatko, and A. Konski, Phase I trial of preoperative hypofractionated
intensity-modulated radiotherapy with incorporated boost and oral
capecitabine in locally advanced rectal cancer, Int. J. Radiat. Oncol.,
Biol., Phys. 675, 13891393 2007.
61
T. Iuchi, K. Hatano, Y. Narita, T. Kodama, T. Yamaki, and K. Osato,
Hypofractionated high-dose irradiation for the treatment of malignant
astrocytomas using simultaneous integrated boost technique by IMRT,
5240
5240
86
5241
5241
radiation dose calculation, IEEE Symposium on Field-Programmable
custom Computing Machines FCCM April 2006.
112
C. C. Ling, J. Humm, S. Larson, H. Amols, Z. Fuks, S. Leibel, and J. A.
Koutcher, Towards multidimensional radiotherapy MD-CRT: Biological imaging and biological conformality, Int. J. Radiat. Oncol., Biol.,
Phys. 473, 551560 2000.
Christian P. Karger
Department of Medical Physics in Radiation Oncology, German Cancer Research Center,
Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
Jrgen Debus
Department of Radiation Oncology and Radiotherapy, University Clinic Heidelberg, Heidelberg, Germany
and Heidelberg Ion Beam Therapy Center (HIT), Heidelberg, Germany
Received 5 February 2008; revised 19 September 2008; accepted for publication 22 September 2008;
published 18 November 2008
Currently, there is an increasing interest in heavy ion radiotherapy RT and a number of new
facilities are being installed in Europe and Japan. This development is accompanied by intensive
technical, physical, and clinical research. The authors identify six research fields where progress is
likely and propose a thesis on the expected achievements for each of the fields: 1 Synchrotrons
with active energy variation and three-dimensional beam scanning will be the standard in ion beam
RT. 2 Common standards for precise measurement, prescription, and reporting of dose will be
available. 3 Intensity-modulated particle therapy will be state-of-the-art. 4 Time-adaptive treatments of moving targets will be feasible. 5 Therapeutic effectiveness of heavy ions will be known
for the most important indications while cost effectiveness will remain to be shown. 6 The
potential of high-linear energy transfer radiation will be known. The rationale for each of these
theses is described. 2008 American Association of Physicists in Medicine.
DOI: 10.1118/1.3002307
Key words: radiotherapy, heavy ions, heavy charged particles, relative biological effectiveness
RBE, high-LET
I. BACKGROUND
The interest in heavy ion radiotherapy RT is currently increasing worldwide.14 Similar to protons, heavy ions exhibit
an inverted depth dose profile, the so-called Bragg curve,
which concentrates the dose at the end of the particle range.
By superimposing several monoenergetic Bragg curves, the
Bragg peak can be spreadout to the extension of the tumor in
depth, while still preserving an advantageous dose ratio between tumor and normal tissue. The degree of conformality
of charged particles was further enhanced by introducing active scanning techniques, which allow dose conformation not
only at the distal but also at the proximal edge of the tumor
for each field.5
As the linear energy transfer LET of heavy ions increases significantly towards the end of their range, heavy
ions are biologically more effective in the peak- than in the
plateau region, if the same dose is applied to the same tissue.
This behavior is considered by introducing the relative biological effectiveness RBE, which is defined as the ratio of a
photon dose to the corresponding iso-effective ion dose.5
Therefore, dose has to be prescribed in terms of biologically
equivalent dose =absorbed dose RBE rather than absorbed dose. It is the difference of the increased RBE between the peak and the plateau regions which potentially
increases the therapeutic effectiveness of heavy ion therapy
relative to proton therapy.
5653
0094-2405/2008/3512/5653/11/$23.00
5653
5654
5654
FIG. 2. For passive beam delivery a the dose distribution is shaped by four
components: the range shifter, the modulator wheel or alternatively a ridge
filter, the collimator, and the compensator. The modulation depth is constant over the tumor cross section. For active beam delivery b, a monoenergetic pencil beam is scanned over the tumor cross section. After one slice
is irradiated the energy of the beam is switched actively or passively, in
case of a range shifter to the next energy. Reprinted from Ref. 4 with
permission of Springer.
It should be noted that a more flexible beam delivery system based on the elements of passive systems has been developed and is explored at HIMAC. This so-called layerstacking method10 uses a sequence of different modulators
each with only a small modulation depth, which are then
stacked in depth to achieve the desired depth dose distribution. A multileaf collimator is used to adapt the field size to
the target in each layer. This method leads to a much better
conformation of the dose to the target volume, similar to a
scanning system.
To efficiently measure dose distributions generated with
the dynamic beam scanning system, multichannel detector
systems are used rather than single ion chamber measurements, which are a standard for passive techniques. Although
the dosimetry of heavy ion beams was already harmonized
by the introduction of the Code of Practice published by the
International Atomic Energy Agency TRS-39817, the overall uncertainty of dose determination in ion beams is still
considerably larger than in conventional radiotherapy. Moreover, there is no common standard for the reporting of biological effective dose and RBE in ion beam therapy.
For a passively modulated beam, the variation of the RBE
with depth has to be taken into account in the design of the
5655
ridge filters or modulators i.e., the hardware. With the selection of a certain modulator, the depth modulation, and
hence, the RBE profile is thus fixed. As the RBE profile
depends not only on the treatment schedule e.g., dose per
fraction, number of daily applied fields but also the type of
tumor and normal tissue or the optimization technique multiple fields versus single field optimization, the flexibility of
passive systems is limited. The realization of an intensity
modulated particle beam therapy IMPT was also never realized with a passive beam application system although in
principle the above mentioned layer-stacking system offers
this possibility.
While it is the current standard in ion beam therapy to
detect interfractional movements of bony structures or metal
markers by orthogonal x rays and to correct for thereafter,
the management of intrafractional movement is more difficult. Due to the quasistatic treatment fields obtained with
passive beam shaping techniques, the treatment of moving
organs is not much different from that in conventional radiotherapy with photons. It has been shown at HIMAC that
respiratory gating of the beam, e.g., by optical motion detection, can be used for ion beam therapy.18 The only additional
problem is the increased treatment time due to the pulsed
beam. For the current beam scanning systems, however, the
treatment of moving organs is a much greater challenge due
to interplay effects between organ motion and the scanned
beam for a detailed discussion see Ref. 19.
The clinical experience gained with ion beams so far is
very limited. The effectiveness of ion beams was demonstrated only for a limited number of tumors and with limited
patient numbers an overview of clinical data is found in
Refs. 9 and 2022. Moreover, the potential advantages of
high LET radiation versus low LET could only be demonstrated by comparison with historical clinical results and not
by directly comparing proton and ion beams in the same
setting. The available clinical data are mainly obtained using
carbon ions and are very scarce for other high LET ions.8
5655
ning for ion beams the standard technique for all new
clinical ion beam facilities. Furthermore, the development of gantry systems for ions will continue and at
least some new facilities will install gantries.
Dosimetry: Common standards for precise measurement, prescription, and reporting of dose will be
available.
The overall uncertainty in determining the absorbed
dose to water in an ion beam will be at the same level as
for megavoltage x rays today. The main improvements
will come from a better knowledge of the underlying
physical parameters. Water calorimetry will serve as reference dosimetry and multichannel dosimetry systems
will facilitate the measurement of two-dimensional 2D
and 3D dose distribution. There will be standardized
methods to report the biological effective dose and the
RBE.
Treatment planning: Intensity-modulated particle
therapy will be state-of-the-art.
The advancements of IMRT in conventional and proton radiotherapy will immediately be introduced in ion
beam therapy. The principles of IMPT based on the optimization of biological effective dose have been developed and, due to the rapid increase of computing power,
will be incorporated in any treatment planning system
for ion beams. IMPT can and will further improve the
conformality of the applied dose distributions. Since
beam delivery by active beam scanning is intrinsically
controlling and modulating the intensity, IMPT will be
much more straightforward to be implemented as it was
for conventional RT with open fields and therefore requires little to no additional effort for quality assurance.
Moving organs: Time-adaptive treatments of moving targets will be feasible.
Although the principal problem is the same for photons and ions, it has to be considered that organ movements do not only change the lateral position of the target, but also its radiological depth, and hence, the
required range of the ions. For heavy ions, changes in
the depth modulation may also require a repeated optimization of the biologically effective dose. In the future,
interfractional movements will be detected by on-board
imaging and will be compensated by setup corrections
and if necessary also by adaptation of the treatment plan.
Intrafractional organ movements will be monitored in
real time and will be compensated to some extent by
using gating or tracking techniques. Improved imaging
technology like on-board cone beam computed tomography CT will also allow direct visualization of the
tumor and soft tissue structures, rather than just bony
structures or markers.
Clinical application: The therapeutic effectiveness of
heavy ions will be known for the most important indications. The cost effectiveness of heavy ion therapy,
however, will still have to be shown.
With the upcoming of new heavy ion centers, the
most important question is related to the safety and effectiveness of this new treatment modality. Both aspects
5656
Two new designs of accelerators are currently being discussed, which would be more compact, and hence, cheaper
than current solutions.
The first one uses laser induced acceleration of ions. Here,
a very high-intensity laser incidents on a thin metal target
and causes ionization. An electromagnetic shockwave causes
electrons to be accelerated in the target, which then ionize
the molecules on the backsurface of the target. The cloud of
electrons is then emitted from the backsurface, causing a
very strong electrostatic field 1013 V m1 that accelerates
protons and ions from the backlayer of the target thickness
Medical Physics, Vol. 35, No. 12, December 2008
5656
5657
5657
5658
5658
FIG. 3. Distribution of the biological effective dose for the irradiation of a skull base tumour, using two nearly opposing lateral fields of carbon ion ions. The
treatment plans on the left and right side were generated with a separate and a simultaneous optimization of the dose distributions in both fields, respectively.
The target volume thick line is shown together with the isodose lines at 90%, 80%, 70%, 50%, 30%, and 10% of the prescribed dose, respectively.
Simultaneous optimization of multiple fields is an important feature of IMPT and its technical realization is in principle just a numerical problem, since the particle numbers of
all beam spots of all fields have to be optimized at the same
time, rather than separately. Due to the large number of beam
spots per field typically 50 000 for a skull base tumor, the
required computer memory and computing time is currently
considerably typically 1.2 GB and 4000 s on a 1.2 GHz
Power4-CPU, respectively. With the expected increase in
computing power, however, this will not be a serious restriction in the near future. IMPT has already been clinically
applied at GSI with the existing scanning technology and
without additional efforts for the quality assurance.40,41 Starting from this basis, clinical application of IMPT will continuously be developed. Figure 3 shows an example of an
IMPT plan including optimization of biological effective
doses.
Due to the sharp dose gradients especially at the distal end
of the Bragg peak, setup errors, organ movements, and range
uncertainties may have significant impact on the quality of
the dose distributions. Resulting over- and undershooting has
to be considered by adequate safety margins, which may be
determined, e.g., by simulated target point displacements or
modified CT-number/range-calibration curves. By introducing IMPT, it has been shown that setup errors do not lead to
worse treatment plans than without IMPT and therefore, robustness against setup-related range uncertainties will be included as an additional constraint in the dose optimization
process.
Since the number of fields applied in ion beam therapy is
generally smaller than in photon IMRT, the selection of
proper beam angles becomes more important. As the range
Medical Physics, Vol. 35, No. 12, December 2008
5659
5659
5660
5660
FIG. 4. Distribution of OER at 10% survival level for V79 and HSG cells
exposed to helium He: , carbon 12C: , or neon 20Ne: ions as a
function of the dose-averaged LET. Reprinted from Ref. 63 with permissions of the author and Radiation Research.
5661
on the cost effectiveness of ion beam therapy become available before solid clinical data on the clinical effectiveness
are available.
IV.F. Radiobiology
5661
5662
Particle Newsletter 36, edited by J. Sisterson Harvard Cyclotron Laboratory, Harvard University, Boston, 2005, www.ptcog.com.
8
J. R. Castro, Results of heavy ion radiotherapy, Radiat. Environ. Biophys. 34, 4548 1995.
9
H. Tsujii et al., Clinical results of carbon ion radiotherapy at NIRS, J.
Radiat. Res. Tokyo 48 Suppl A, A1A13 2007.
10
M. Torikoshi, S. Minohara, N. Kanematsu, M. Komori, M. Kanazawa, K.
Noda, N. Miyahara, H. Itoh, M. Endo, and T. Kanai, Irradiation System
for HIMAC, J. Radiat. Res. Tokyo 48 Suppl A, A15A25 2007.
11
Y. Hishikawa, Y. Oda, H. Mayahara, A. Kawaguchi, K. Kagawa, M.
Murakami, and M. Abe, Status of the clinical work at Hyogo, Radiother. Oncol. 73 Suppl 2, S38S40 2004.
12
P. Heeg, H. Eickhoff, and T. Haberer, Conception of heavy ion beam
therapy at Heidelberg University HICAT, Z. Med. Phys. 14, 1724
2004.
13
T. Haberer, J. Debus, H. Eickhoff, O. Jkel, D. Schulz-Ertner, and U.
Weber, The Heidelberg Ion Therapy Center, Radiother. Oncol. 73
Suppl 2, S186S190 2004.
14
U. Amaldi, CNAOThe Italian Centre for Light-Ion Therapy, Radiother. Oncol. 73 Suppl 2, S191S201 2004.
15
U. Amaldi and G. Kraft, European developments in radiotherapy with
beams of large radiobiological effectiveness, J. Radiat. Res. Tokyo 48
Suppl A, A27A41 2007.
16
Q. Li, Z. Dai, Z. Yan, X. Jin, X. Liu, and G. Xiao, Heavy-ion conformal
irradiation in the shallow-seated tumor therapy terminal at HIRFL, Med.
Biol. Eng. Comput. 4511, 10371043 2007.
17
P. Andreo, D. T. Burns, K. Hohlfeld, M. S. Huq, T. Kanai, F. Laitano, V.
Smyth, and S. Vynckier, Absorbed dose determination in external beam
radiotherapy, Technical Report Series No. 398 2000.
18
S. Minohara, T. Kanai, M. Endo, K. Noda, and M. Kanazawa, Respiratory gated irradiation system for heavy-ion radiotherapy, Int. J. Radiat.
Oncol., Biol., Phys. 47, 10971103 2000.
19
C. Bert, N. Saito, A. Schmidt, N. Chaudhri, D. Schardt, and E. Rietzel,
Target motion tracking with a scanned particle beam, Med. Phys. 34,
47684771 2007.
20
D. Schulz-Ertner, A. Nikoghosyan, H. Hof, B. Didinger, S. E. Combs, O.
Jkel, C. P. Karger, L. Edler, and J. Debus, Carbon ion radiotherapy of
skull base chondrosarcomas, Int. J. Radiat. Oncol., Biol., Phys. 67, 171
177 2007.
21
D. Schulz-Ertner, C. P. Karger, A. Feuerhake, A. Nikoghosyan, S. E.
Combs, O. Jkel, L. Edler, M. Scholz, and J. Debus, Effectiveness of
carbon ion radiotherapy in the treatment of skull-base chordomas, Int. J.
Radiat. Oncol., Biol., Phys. 68, 449457 2007.
22
D. Schulz-Ertner and H. Tsujii, Particle radiation therapy using proton
and heavier ion beams, J. Clin. Oncol. 25, 953964 2007.
23
D. Schulz-Ertner, A. Nikoghosyan, B. Didinger, M. Munter, O. Jkel, C.
P. Karger, and J. Debus, Therapy strategies for locally advanced adenoid
cystic carcinomas using modern radiation therapy techniques, Cancer
104, 338344 2005.
24
K. W. Ledingham, W. Galster, and R. Sauerbrey, Laser-driven proton
oncologyA unique new cancer therapy?, Br. J. Radiol. 80, 855858
2007.
25
G. J. Caporaso, 1994 US-CERN-Japan Joint Accelerator School: Topical
Course: Frontiers Of Accelerator Technology, 39 Nov 1994, Maui, Hawaii, edited by M. M. S. T. S. I. Kurokawa World Scientific, New Jersey,
1996, pp. 594615.
26
S. E. Sampayan, G. J. Caporaso, D. M. Sanders, R. D. Stoddard, D. O.
Trimble, J. Elizondo, M. L. Krogh, and T. F. Wieskamp, Particle Accelerator Conference, 1997. Proceedings of the 1997 IEEE, Lawrence Livermore Nat. Lab., 1997, pp. 13081310.
27
Y.-J. Chen and A. C. Paul, Proceedings of PAC07, Albuquerque, New
Mexico, USA, edited by S. Hardage and C. Petit-Jean-Genaz IEEE, Piscataway, NJ, 2007, pp. 17871789.
28
D. Trbojevic, D. Parker, E. Keil, and A. M. Sessler, Carbon/proton
therapy: A novel gantry design, Phys. Rev. ST Accel. Beams 2007,
053503-1053503-6 2007.
29
Y. Jongen et al., Design Studies of the Compact Superconducting Cyclotron for Hadron Therapy, Abstract TUPLS078, Proceedings of EPAC,
Edinburgh, Scotland, 16781680 2006.
30
H. Palmans, J. Seuntjens, F. Verhaegen, J. M. Denis, S. Vynckier, and H.
Thierens, Water calorimetry and ionization chamber dosimetry in an
85-MeV clinical proton beam, Med. Phys. 23, 643650 1996.
31
H. J. Brede, K. D. Greif, O. Hecker, P. Heeg, J. Heese, D. T. Jones, H.
7
5662
Kluge, and D. Schardt, Absorbed dose to water determination with ionization chamber dosimetry and calorimetry in restricted neutron, photon,
proton and heavy-ion radiation fields, Phys. Med. Biol. 51, 36673682
2006.
32
F. Sauli, GEM: A new concept for electron amplification in gas detectors, Nucl. Instrum. Methods Phys. Res. A 386, 531534 1997.
33
G. T. Y. Chen, S. Mori, A. Chu, and M. Folkert, Uncertainties in image
guided radiotherapy of lung tumors, Proceedings of NIRS-CNAO Joint
Symposium on Carbon Ion Radiotherapy, Milan, Italy, 2006, pp. 192193
2006.
34
C. Zhang, P. Lechner, G. Lutz, M. Porroe, R. Richter, J. Treisa, L.
Strder, and S. N. Hang, Development of DEPFET Macropixel detectors, Nucl. Instrum. Methods Phys. Res. A 568, 207216 2008.
35
J. Jakubek, T. Holy, E. Lehmann, S. Pospisil, J. Uher, D. Vacik, and D.
Vavrik, Spatial resolution of Medipix-2 device as neutron pixel detector, Nucl. Instrum. Methods Phys. Res. A 546, 164169 2005.
36
J. Jakubek et al., Neutron imaging and tomography with Medipix2 and
dental micro-roentgenography, Nucl. Instrum. Methods Phys. Res. A
569, 205209 2006.
37
M. Rebisz, B. Voss, A. Heinz, E. Usenko, and M. Pomorski, CVD diamond dosimeters for heavy ion beams, Diamond Relat. Mater. 16, 1070
1073 2007.
38
A. Wambersie, J. H. Hendry, P. Andreo, P. M. DeLuca, R. Gahbauer, H.
Menzel, and G. Whitmore, The RBE issues in ion-beam therapy: Conclusions of a joint IAEA/ICRU working group regarding quantities and
units, Radiat. Prot. Dosim. 122, 463470 2006.
39
M. Krmer and M. Scholz, Rapid calculation of biological effects in ion
radiotherapy, Phys. Med. Biol. 51, 19591970 2006.
40
M. Ellerbrock, O. Jkel, M. Krmer, A. Nikoghosyan, and D. SchulzErtner, Clinical implementation of intensity modulated heavy ion
therapy, Radiother. Oncol. 84, 129 2007.
41
M. Ellerbrock, O. Jkel, M. Krmer, A. Nikoghosyan, and D. SchulzErtner, Multiple field optimization in heavy ion treatment planning
clinical aspects, in Abstracts der Frhjahrstagung in Heidelberg Deutsche Physikalische Gesellschaft DPG ST 6.4, Heidelberg, 2006.
42
M. Engelsman, E. Rietzel, and H. M. Kooy, Four-dimensional proton
treatment planning for lung tumors, Int. J. Radiat. Oncol., Biol., Phys.
64, 15891595 2006.
43
E. Rietzel, S. J. Rosenthal, D. P. Gierga, C. G. Willet, and G. T. Chen,
Moving targets: Detection and tracking of internal organ motion for
treatment planning and patient set-up, Radiother. Oncol. 73 Suppl 2,
S68S72 2004.
44
S. Mori, G. T. Chen, and M. Endo, Effects of intrafractional motion on
water equivalent pathlength in respiratory-gated heavy charged particle
beam radiotherapy, Int. J. Radiat. Oncol., Biol., Phys. 69, 308317
2007.
45
P. Kupelian, T. Willoughby, A. Mahadevan, T. Djemil, G. Weinstein, S.
Jani, C. Enke, T. Solberg, N. Flores, D. Liu, D. Beyer, and L. Levine,
Multi-institutional clinical experience with the Calypso System in localization and continuous, real-time monitoring of the prostate gland during
external radiotherapy, Int. J. Radiat. Oncol., Biol., Phys. 67, 10881098
2007.
46
D. W. Litzenberg, J. M. Balter, S. W. Hadley, H. M. Sandler, T. R.
Willoughby, P. A. Kupelian, and L. Levine, Influence of intrafraction
motion on margins for prostate radiotherapy, Int. J. Radiat. Oncol., Biol.,
Phys. 65, 548553 2006.
47
ICRU Report 62, Prescribing, Recording and Reporting Photon Beam
Therapy supplement to ICRU report 50 International Commission on
Radiation Units and Measurements, Washington, DC, 1999.
48
S. Minohara, M. Endo, T. Kanai, H. Kato, and H. Tsujii, Estimating
uncertainties of the geometrical range of particle radiotherapy during respiration, Int. J. Radiat. Oncol., Biol., Phys. 56, 121125 2003.
49
C. Bert, S. O. Grozinger, and E. Rietzel, Quantification of interplay
effects of scanned particle beams and moving targets, Phys. Med. Biol.
53, 22532265 2008.
50
T. Furukawa, T. Inaniwa, S. Sato, T. Tomitani, S. Minohara, K. Noda, and
T. Kanai, Design study of a raster scanning system for moving target
irradiation in heavy-ion radiotherapy, Med. Phys. 34, 10851097 2007.
51
C. Bert and E. Rietzel, 4D treatment planning for scanned ion beams,
Radiother. Oncol. 2, 24 2007.
52
S. O. Grozinger, E. Rietzel, Q. Li, C. Bert, T. Haberer, and G. Kraft,
Simulations to design an online motion compensation system for
scanned particle beams, Phys. Med. Biol. 51, 35173531 2006.
5663
53
5663
Kevin Cleary
Imaging Science and Information Systems, Georgetown University Hospital, Washington, DC 20007
Warren DSouza
Radiation Oncology, University of Maryland, College Park, Maryland 20742
Martin Murphy
Radiation Oncology, Virginia Commonwealth University, Richmond, Virginia 23284
Kenneth H. Wong
Imaging Science and Information Systems, Georgetown University Hospital, Washington, DC 20007
Paul Kealla
Radiation Oncology, Stanford University, Stanford, California 94305
Received 18 March 2008; revised 17 October 2008; accepted for publication 17 October 2008;
published 19 November 2008
The current climate of rapid technological evolution is reflected in newer and better methods to
modulate and direct radiation beams for cancer therapy. This Vision 20/ 20 paper focuses on part of
this evolution, locating and targeting moving tumors. The two processes are somewhat independent
and in principle different implementations of the locating and targeting processes can be interchanged. Advanced localization and targeting methods have an impact on treatment planning and
also present new challenges for quality assurance QA, that of verifying real-time delivery. Some
methods to locate and target moving tumors with radiation beams are currently FDA approved for
clinical useand this availability and implementation will increase with time. Extensions of current
capabilities will be the integration of higher order dimensionality, such as rotation and deformation
in addition to translation, into the estimate of the patient pose and real-time reoptimization and
adaption of delivery to the dynamically changing anatomy of cancer patients. 2008 American
Association of Physicists in Medicine. DOI: 10.1118/1.3020593
Key words: respiratory motion, adaptive radiotherapy, tracking
I. INTRODUCTION
This Vision 20/ 20 paper focuses on the evolution of locating
and targeting moving tumors. The rationale for using this
technology in the clinic is to significantly improve tumor
control while reducing critical organ damage for tumors
that move with respiration, resulting in a better quality of
life, or beneficence for cancer patients. Beneficence is
one of the three basic principles of the Belmont
report, http://www.hhs.gov/ohrp/humansubjects/guidance/
belmont.htm#xbenefit one of the most important documents
in the field of medical ethics. This report states The term
beneficence is often understood to cover acts of kindness
or charity that go beyond strict obligation. In this document,
beneficence is understood in a stronger sense, as an obligation. Two general rules have been formulated as complementary expressions of beneficent actions in this sense: (1) Do
not harm and (2) maximize possible benefits and minimize
possible harms. Beneficence, as it applies to image-guided
therapy, can be achieved by reducing the difference between
the planned and delivered dose, reducing treatment uncertainties and therefore margins, and improving dose conformation to the tumor. Our patients and the treatment team
should be demanding this beneficence. As with many
5684
0094-2405/2008/3512/5684/11/$23.00
5684
5685
Dieterich et al.: Locating and targeting moving tumors with radiation beams
5685
Treatment
unit
Correct for
reaction time
Treatment
beam
Detect
motion
Moving
Target (tumor)
FIG. 1. Observed variation in lung tumor motion with time in three dimensions from Cyberknife Synchrony data. Ref. 113 Notable is that there is
significant motion in three dimensions, there are variations in the baseline
position of the tumor in all three dimensions and variations in the cycle-tocycle period, respiratory cycle shape, and range of motion.
5686
Dieterich et al.: Locating and targeting moving tumors with radiation beams
5686
Data dimensionality
Invasive?
Availability
Imaging frequency
Points
Points
Points
Points
Volume
Volumes
Yes
Yes
Noa and Yes
Yes
No
No
10 Hz
30 Hz
Before every beam/30 Hz
30 Hz theoretical
N/A
25 Hz
with much less information than a full 4D CT. Finally, although on-line target localization is appealing from the technical point of view and may produce clinical benefits in
some cases, the ultimate test of its value will be its impact on
patient care.
II.B. Methods for target tracking
The heart of a real-time tumor tracking system is the control loop that connects the tumor detection system with the
targeting alignment system. The control loop must receive
target coordinate measurements, test that the target identification is authentic so as not to go off chasing false motion,
filter the target data to avoid disruptive transients, and then
send the delivery system a prediction of where the tumor will
be when the targeting adjustment has been completed. Tumor
tracking requires motors that can guide the beam or couch
faster than most respiratory motion and a fast secondary
feedback mechanism for each motor used to control the
beam or patient placement. Table II compares several motion tracking methods.
The various motion adaptation systems in use and under
consideration involve two fundamentally different types of
control loop. The CyberKnife robotic treatment system,2629
the multi-leaf collimator,3042 gimbaled linac,43 and proton
and heavy ion treatment systems44,45 all use an open-loop
control design. In an open loop system, the corrective response has no effect on the target behavior that signaled the
response. In other words, there is no feedback between the
corrective signal and the adaptive response. In contrast, a
couch compensation system4649 has a closed-loop control
system. When the tumor moves within the patient, the detection system triggers a compensating movement of the patient
Tracking method
Block Ref. 112
Couch Refs. 4648
DMLC Refs. 3040
Gimbaled linac Ref. 43
Magnetic/mechanical particle beam Refs. 44 and 45
Robotic linac Refs. 2629
Approximate system
delay ms
Translation
N/A
Translation, Rotation
Translation, Rotation, Deformation
Translation, Rotation, Deformation
Translation, Rotation, Deformation
Translation, Rotation, Deformation
80
160
N/A
80
110
Availability
Translated to patient treatments
at one center
Under development
Under development
Under development
Under development
Commercially available
5687
Dieterich et al.: Locating and targeting moving tumors with radiation beams
and thus the tumor via the treatment couch. Thus, there is
feedback through which the adaptive response may influence
the target movement.
Real-time systems must react to change at least as fast as
it occurs. A systems reaction time depends on a number of
factors. Time is required to detect a change in the target
position, process and filter the position data, communicate
with the beam alignment system, and complete the repositioning of either the beam or patient. If a single motion detection method fluoroscopic imaging, for example is used
in a first order open loop system, then all of these time delays
combine linearly in series to give the total reaction time. On
the other hand, if two different motion detection systems are
used in parallel to provide partially redundant target position
data, then only the faster systems delay adds into the reaction time of an open loop system. For example, the CyberKnife uses an optical tracking system to provide fast, continuous estimates of a tumors respiratory induced motion,
with the estimate updated by periodic x-ray imaging.27 This
is an example of a hybrid detection method. The total system
reaction time includes the optical processing time while the
time spent processing the x-ray images can be done outside
the duty cycle of the control loop. This allows the targeting
system to invest more time in sophisticated image processing
without a corresponding degradation of response time.
Closed loop systems such as a movable couch can have
higher order dynamics that involve multiple system response
time constants in addition to the dead time before the couch
begins its corrective response.46 In an open loop system the
total reaction time must be compensated by predicting the
future position of the target so that the beam reaches the
future target position at the same time as the tumor. A closed
control loop needs to predict the dead time before the system
begins to respond to a change in the tumor position.46
The future displacement of a breathing motion can be
predicted in several ways: 1 With a mathematical model of
the breathing signal,50 2 with a bio-mechanical model of
the breathing process,51 or 3 with heuristic learning algorithms that mimic the breathing process as it is observed.52,53
The last category includes neural network prediction algorithms, which have been studied by several groups.5255 Neural networks can adapt to changes in the breathing pattern as
they occur, making them an attractive way to deal with the
nonstationary character of real breathing.
II.C. Treatment planning
5687
5688
Dieterich et al.: Locating and targeting moving tumors with radiation beams
5688
tion path of the tumor. The problem is formulated as a standard linear program and solved by minimizing the total
monitor units.
The most widely reported 4D planning method involves
the coregistration of dose distributions calculated on individual 3D CT data sets corresponding to various phases of
the respiration cycle with a single reference 3D CT data set
typically end-inhale or end-exhale.6468,7177 This approach
relies on the image registration algorithm to provide displacement vectors that show the trajectory of each voxel
from one image set to another. The dose distribution calculated on each CT data set is then weighted by the timeweight of the CT phase in the respiration cycle and accumulated in the reference image. In this way one can see
differences between the 4D computed dose and the dose calculated on individual 3D CT data. This method has been
applied for 4D proton treatment planning as well.78,79
Future developments in real-time control will aim to reduce system reaction time Table II while improving temporal prediction to compensate for delays. Reaction time can be
reduced by increasing the speed of each stage of target acquisition, signal processing, communication, and adaptive
alignment response. It can also be reduced by developing
5689
Dieterich et al.: Locating and targeting moving tumors with radiation beams
5689
The complexity of these real-time motion adaptive technologies has made it very difficult for a single person to
understand in detail where potential failures might occur.
Current pathways to disseminate QA information are slow
compared to the speed at which new products are implemented in clinical practice. However, procedures and equipment need to be evaluated thoroughly in a clinical setting to
ensure that a comprehensive quality control QC and QA
program can be developed. The knowledge of experts in the
field of safety such as industrial process engineers has
scarcely been tapped in the health care environment.
The traditional QA approach had drawbacks in the age of
rapidly developing, complex technology such as real-time
motion adaptive radiation therapy. As a first step, the use of
nonstationary phantoms which simulate skin and tumor motion separately is becoming a standard tool in the QA process
to measure the precision and accuracy of respiratory motion
compensation. The major limitation of these phantoms is the
dependency of the measurement result on the simulated motion pattern used. Because we still do not have ground
truth data of real-time tumor motion and surrogate marker
for the time intervals required for treatment maximum 150 s
of data vs. 15 60 min treatment length, the results will not
truly reflect patient treatment results. In addition, simulating
rotation and deformation is still challenging. A minor, but
significant, issue may be the effects on the dose distribution
due to the change of the dose calculation parameters
throughout the respiratory cycle e.g., SSD, surface correction, density changes of lung. This potential change of dose
distribution can only be simulated and measured in anthropomorphic phantoms or very sophisticated software which
takes into account the body deformation in correlation with
the timing of the respective beam delivery with the corresponding respiratory phase.
Accuracy measurements with this new generation of
phantoms will give the medical physicist the answer to the
question of technical accuracy of the real-time motion adaptive treatment delivery system.93 Indirect tracking methods
such as hybrid skin/tumor tracking models and/or 4DCT
based simulations of the correlation between internal motion
and a surrogate respiratory marker do not provide real-time
position information. This in turn introduces an additional
error, namely the uncertainly from the correlation model between external surrogate and tumor position.29 Phantoms are
not sufficiently anthropomorphic, and real-time in vivo dosimetry can so far only provide a few checkpoints of the
delivery accuracy of a 3D treatment plan.
As treatments move towards real-time feedback and realtime adaptation, QA processes will need to account for the
increased delivery complexity and shorter timescales of
beam adjustment. This will involve the addition of real-time
measurement as appropriate on the radiation delivery devices
themselves and synchronization of measured motion and
dose delivery data streams to facilitate dose reconstruction to
estimate the dose delivered to the patient. Also, phantoms
5690
Dieterich et al.: Locating and targeting moving tumors with radiation beams
5690
5691
Dieterich et al.: Locating and targeting moving tumors with radiation beams
5691
Lagendijk, Integrating a MRI scanner with a 6 MV radiotherapy accelerator: Dose deposition in a transverse magnetic field, Phys. Med. Biol.
49, 41094118 2004.
11
J. J. Lagendijk, B. W. Raaymakers, A. J. Raaijmakers, J. Overweg, K. J.
Brown, E. M. Kerkhof, R. W. van der Put, B. Hardemark, M. van Vulpen,
and U. A. van der Heide, MRI/linac integration, Radiother. Oncol. 86,
2529 2008.
12
A. J. Raaijmakers, B. W. Raaymakers, and J. J. Lagendijk, Experimental
verification of magnetic field dose effects for the MRI-accelerator, Phys.
Med. Biol. 52, 42834291 2007.
13
C. Kirkby, T. Stanescu, S. Rathee, M. Carlone, B. Murray, and B. G.
Fallone, Patient dosimetry for hybrid MRI-radiotherapy systems, Med.
Phys. 35, 10191027 2008.
14
A. Hsu, N. R. Miller, P. M. Evans, J. C. Bamber, and S. Webb, Feasibility of using ultrasound for real-time tracking during radiotherapy,
Med. Phys. 32, 15001512 2005.
15
J. Wu, O. Dandekar, D. Nazareth, P. Lei, W. DSouza, and R. Shekhar,
Effect of ultrasound probe on dose delivery during real-time ultrasoundguided tumor tracking, Conf. Proc. IEEE Eng. Med. Biol. Soc. 1, 3799
3802 2006.
16
A. Sawada, K. Yoda, M. Kokubo, T. Kunieda, Y. Nagata, and M. Hiraoka,
A technique for noninvasive respiratory gated radiation treatment system
based on a real time 3D ultrasound image correlation: A phantom study,
Med. Phys. 31, 245250 2004.
17
M. Fuss, J. Boda-Heggemann, N. Papanikolau, and B. J. Salter, Imageguidance for stereotactic body radiation therapy, Med. Dosim. 32, 102
110 2007.
18
T. R. Willoughby, P. A. Kupelian, J. Pouliot, K. Shinohara, M. Aubin, M.
Roach, 3rd, L. L. Skrumeda, J. M. Balter, D. W. Litzenberg, S. W. Hadley, J. T. Wei, and H. M. Sandler, Target localization and real-time
tracking using the Calypso 4D localization system in patients with localized prostate cancer, Int. J. Radiat. Oncol., Biol., Phys. 65, 528534
2006.
19
J. M. Balter, J. N. Wright, L. J. Newell, B. Friemel, S. Dimmer, Y. Cheng,
J. Wong, E. Vertatschitsch, and T. P. Mate, Accuracy of a wireless localization system for radiotherapy, Int. J. Radiat. Oncol., Biol., Phys. 61,
933937 2005.
20
P. Kupelian, T. Willoughby, A. Mahadevan, T. Djemil, G. Weinstein, S.
Jani, C. Enke, T. Solberg, N. Flores, D. Liu, D. Beyer, and L. Levine,
Multi-institutional clinical experience with the Calypso System in localization and continuous, real-time monitoring of the prostate gland during
external radiotherapy, Int. J. Radiat. Oncol., Biol., Phys. 67, 10881098
2007.
21
L. Dietrich, S. Jetter, T. Tucking, S. Nill, and U. Oelfke, Linacintegrated 4D cone beam CT: First experimental results, Phys. Med.
Biol. 51, 29392952 2006.
22
N. Wink, C. Panknin, and T. D. Solberg, Phase versus amplitude sorting
of 4D-CT data, Med. Phys. 7, 7785 2006.
23
J. W. Wong, M. B. Sharpe, D. A. Jaffray, V. R. Kini, J. M. Robertson, J.
S. Stromberg, and A. A. Martinez, The use of active breathing control
ABC to reduce margin for breathing motion, Int. J. Radiat. Oncol.,
Biol., Phys. 44, 911919 1999.
24
J. Hanley, M. M. Debois, D. Mah, G. S. Mageras, A. Raben, K. Rosenzweig, B. Mychalczak, L. H. Schwartz, P. J. Gloeggler, W. Lutz, C. C.
Ling, S. A. Leibel, Z. Fuks, and G. J. Kutcher, Deep inspiration breathhold technique for lung tumors: The potential value of target immobilization and reduced lung density in dose escalation, Int. J. Radiat. Oncol.,
Biol., Phys. 45, 603611 1999.
25
S. S. Korreman, T. Juhler-Nottrup, and A. L. Boyer, Respiratory gated
beam delivery cannot facilitate margin reduction, unless combined with
respiratory correlated image guidance, Radiother. Oncol. 86, 6168
2008.
26
M. J. Murphy, Tracking moving organs in real time, Semin. Radiat.
Oncol. 14, 91100 2004.
27
A. Schweikard, H. Shiomi, and J. Adler, Respiration tracking in radiosurgery, Med. Phys. 31, 27382741 2004.
28
C. Ozhasoglu, M. J. Murphy, G. Glosser, M. Bodduluri, A. Schweikard,
K. Forster, D. P. Martin, and J. R. Adler, Real-time tracking of the tumor
volume in precision radiotherapy and body radiosurgeryA novel approach to compensate for respiratory motion, presented at the Computer
Assisted Radiology and Surgery, San Francisco, 2000.
5692
Dieterich et al.: Locating and targeting moving tumors with radiation beams
29
5692
5693
Dieterich et al.: Locating and targeting moving tumors with radiation beams
71
94
5693
N. C. Choi and J. A. Doucette, Improved survival of patients with unresectable non-small-cell bronchogenic carcinoma by an innovated highdose en-bloc radiotherapeutic approach, Cancer 48, 101109 1981.
95
M. K. Martel, R. K. Ten Haken, M. B. Hazuka, M. L. Kessler, M. Strawderman, A. T. Turrisi, T. S. Lawrence, B. A. Fraass, and A. S. Lichter,
Estimation of tumor control probability model parameters from 3-D dose
distributions of non-small cell lung cancer patients, Lung Cancer 24,
3137 1999.
96
P. Okunieff, D. Morgan, A. Niemierko, and H. D. Suit, Radiation doseresponse of human tumors, Int. J. Radiat. Oncol., Biol., Phys. 32, 1227
1237 1995.
97
C. A. Perez, M. Bauer, S. Edelstein, B. W. Gillespie, and R. Birch, Impact of tumor control on survival in carcinoma of the lung treated with
irradiation, Int. J. Radiat. Oncol., Biol., Phys. 12, 539547 1986.
98
C. A. Perez, T. F. Pajak, P. Rubin, J. R. Simpson, M. Mohiuddin, L. W.
Brady, R. Perez-Tamayo, and M. Rotman, Long-term observations of the
patterns of failure in patients with unresectable non-oat cell carcinoma of
the lung treated with definitive radiotherapy. Report by the Radiation
Therapy Oncology Group, Cancer 59, 18741881 1987.
99
C. A. Perez, K. Stanley, P. Rubin, S. Kramer, L. Brady, R. Perez-Tamayo,
G. S. Brown, J. Concannon, M. Rotman, and H. G. Seydel, A prospective randomized study of various irradiation doses and fractionation
schedules in the treatment of inoperable non-oat-cell carcinoma of the
lung. Preliminary report by the Radiation Therapy Oncology Group,
Cancer 45, 27442753 1980.
100
J. Wulf, K. Baier, and M. P. Flentje, Dose escalation in radiothreapy of
lung tumors by stereotactic irradiation: Is there a dose-response relationship for local tumor control? Int. J. Radiat. Oncol., Biol., Phys. 63, S52
2005.
101
J. Bradley, J. O. Deasy, S. Bentzen, and I. El-Naqa, Dosimetric correlates for acute esophagitis in patients treated with radiotherapy for lung
carcinoma, Int. J. Radiat. Oncol., Biol., Phys. 58, 11061113 2004.
102
J. Bradley, M. V. Graham, K. Winter, J. A. Purdy, R. Komaki, W. H. Roa,
J. K. Ryu, W. Bosch, and B. Emami, Toxicity and outcome results of
RTOG 9311: A phase I-II dose-escalation study using three-dimensional
conformal radiotherapy in patients with inoperable non-small-cell lung
carcinoma, Int. J. Radiat. Oncol., Biol., Phys. 61, 318328 2005.
103
M. V. Graham, J. A. Purdy, B. Emami, W. Harms, W. Bosch, M. A.
Lockett, and C. A. Perez, Clinical dose-volume histogram analysis for
pneumonitis after 3D treatment for non-small cell lung cancer NSCLC,
Int. J. Radiat. Oncol., Biol., Phys. 45, 323329 1999.
104
M. L. Hernando, L. B. Marks, G. C. Bentel, S. M. Zhou, D. Hollis, S. K.
Das, M. Fan, M. T. Munley, T. D. Shafman, M. S. Anscher, and P. A.
Lind, Radiation-induced pulmonary toxicity: A dose-volume histogram
analysis in 201 patients with lung cancer, Int. J. Radiat. Oncol., Biol.,
Phys. 51, 650659 2001.
105
S. L. Kwa, J. V. Lebesque, J. C. Theuws, L. B. Marks, M. T. Munley, G.
Bentel, D. Oetzel, U. Spahn, M. V. Graham, R. E. Drzymala, J. A. Purdy,
A. S. Lichter, M. K. Martel, and R. K. Ten Haken, Radiation pneumonitis as a function of mean lung dose: An analysis of pooled data of 540
patients, Int. J. Radiat. Oncol., Biol., Phys. 42, 19 1998.
106
D. Oetzel, P. Schraube, F. Hensley, G. Sroka-Perez, M. Menke, and M.
Flentje, Estimation of pneumonitis risk in three-dimensional treatment
planning using dose-volume histogram analysis, Int. J. Radiat. Oncol.,
Biol., Phys. 33, 455460 1995.
107
Y. Seppenwoolde, J. V. Lebesque, K. de Jaeger, J. S. Belderbos, L. J.
Boersma, C. Schilstra, G. T. Henning, J. A. Hayman, M. K. Martel, and
R. K. Ten Haken, Comparing different NTCP models that predict the
incidence of radiation pneumonitis, Int. J. Radiat. Oncol., Biol., Phys.
55, 724735 2003.
108
E. D. Yorke, A. Jackson, K. E. Rosenzweig, S. A. Merrick, D. Gabrys, E.
S. Venkatraman, C. M. Burman, S. A. Leibel, and C. C. Ling, Dosevolume factors contributing to the incidence of radiation pneumonitis in
non-small-cell lung cancer patients treated with three-dimensional conformal radiation therapy, Int. J. Radiat. Oncol., Biol., Phys. 54, 329339
2002.
109
S. Hossain, P. Xia, C. Chuang, L. Verhey, A. Gottschalk, G. Mu, and L.
Ma, Simulated real time image guided intrafraction tracking-delivery for
hypofractionated prostate IMRT, Med. Phys. 35, 40414048 2008.
5694
Dieterich et al.: Locating and targeting moving tumors with radiation beams
110
5694
Received 8 August 2008; revised 10 October 2008; accepted for publication 15 October 2008;
published 19 November 2008
Commercially available high-resolution three-dimensional optical imaging modalitiesincluding
confocal microscopy, two-photon microscopy, and optical coherence tomographyhave fundamentally impacted biomedicine. Unfortunately, such tools cannot penetrate biological tissue deeper than
the optical transport mean free path 1 mm in the skin. Photoacoustic tomography, which combines strong optical contrast and high ultrasonic resolution in a single modality, has broken through
this fundamental depth limitation and achieved superdepth high-resolution optical imaging. In
parallel, radio frequency-or microwave-induced thermoacoustic tomography is being actively developed to combine radio frequency or microwave contrast with ultrasonic resolution. In this Vision
20/ 20 article, the prospects of photoacoustic tomography are envisaged in the following aspects:
1 photoacoustic microscopy of optical absorption emerging as a mainstream technology, 2
melanoma detection using photoacoustic microscopy, 3 photoacoustic endoscopy, 4 simultaneous functional and molecular photoacoustic tomography, 5 photoacoustic tomography of gene
expression, 6 Doppler photoacoustic tomography for flow measurement, 7 photoacoustic tomography of metabolic rate of oxygen, 8 photoacoustic mapping of sentinel lymph nodes, 9 multiscale photoacoustic imaging in vivo with common signal origins, 10 simultaneous photoacoustic
and thermoacoustic tomography of the breast, 11 photoacoustic and thermoacoustic tomography
of the brain, and 12 low-background thermoacoustic molecular imaging. 2008 American Association of Physicists in Medicine. DOI: 10.1118/1.3013698
Key words: microscopy, melanoma detection, endoscopy, functional imaging, molecular imaging,
reporter gene imaging, Doppler effect, metabolic rate of oxygen, sentinel lymph nodes, multiscale
imaging, thermoacoustic tomography, breast imaging, brain imaging
I. INTRODUCTION
The field of photoacoustic tomography PAT has grown a
great deal in the past few years. PAT is cross-sectional or
three-dimensional imaging based on the photoacoustic effect.
Alexander Graham Bell first reported on the photoacoustic
effect in 1880. Only recently, however, was PAT developed
as an imaging technology.129 PAT combines high ultrasonic
resolution and strong optical contrast in a single modality,
capable of providing high-resolution structural, functional,19
and molecular30,31 imaging in vivo in optically scattering biological tissue at new depths.
In biological tissues, light transfer is dominated by scattering. The mean free path is on the order of 0.1 mm, and the
transport mean free path is on the order of 1 mm. While the
former measures the frequency of predominantly anisotropic
scattering, the latter assesses the frequency of equivalent isotropic scattering. As a result of scattering, photon propagation transitions from the ballistic regime into the diffusive
regime around one transport mean free path.4 Specifically,
the ballistic regime is within the mean free path, where a
significant number of photons have undergone no scattering.
The quasiballistic regime is from the mean free path to the
transport mean free path, where photons have sustained a
few scattering events but retain a strong memory of the original incidence direction. The quasidiffusive regime is from
the transport mean free path to ten times the transport mean
5758
0094-2405/2008/3512/5758/10/$23.00
5758
5759
5759
transducers placed outside the tissue, producing electric signals. The electric signals are then amplified, digitized, and
transferred to a computer, where an image is formed. PAT
depends on any absorbed photons, either unscattered or scattered, to produce photoacoustic signals as long as the photon
excitation is relaxed thermally. Readers are referred to recent
review and tutorial articles for more details.1,4
The formation of a photoacoustic image, mapping the
photoacoustic source according to the detected sound signals, can be exemplified with the simplest case of pinpointing a single sound source such as thunder in threedimensional 3D space. If the time delay between seeing
lightening and hearing thunder is recorded, one can infer that
the thunder took place on a spherical surface of a radius
determined by the product of the speed of sound and the time
delay. If three such measurements are taken at different locations, the sound source can be accurately triangulated. If
produced by the photoacoustic effect, the sound source can
be imaged by PAT. In most practical cases, the sound source
is spatially distributed rather than localized at a point.
PAT has been implemented in two major forms. One form
is based on a scanning focused ultrasonic transducer. Acoustic focusing in combination with time-resolved detection of
photoacoustic waves directly provides 3D spatial resolution.
Scanning over the tissue produces tomographic images.
Dark-field confocal photoacoustic microscopy belongs to
this category.3335 Dark field means that the light illumination
pattern on the tissue surface is donut shaped with a dark
core; confocal means that the ultrasonic detection shares the
same focus as the light illumination. The other form is based
on an array of unfocused ultrasonic transducers. This
method, also referred to as photoacoustic computed tomography, requires the use of an inverse algorithm to reconstruct
a tomographic image.36
The dominant contrast of interest is based on the optical
absorption in the photoacoustic excitation phase. By using
multiwavelength measurement, one can simultaneously
quantify concentrations of multiple chromophores of different colors, such as oxygenated and deoxygenated hemoglobin molecules in red blood cells. Such quantification of hemoglobin can provide functional imaging of the
concentration and oxygen saturation of hemoglobin. Both
parameters are related to hallmarks of cancer: the concentration of hemoglobin correlates with angiogenesis,27 whereas
the oxygen saturation of hemoglobin correlates with
hypermetabolism.31 Such parameters of hemoglobin can also
be used to image brain activity.19 In addition, extrinsic optical absorption contrast agents can be used to provide molecular imaging of biomarkers.30,31
PAT is highly sensitive to optical absorption. PAT works
on dark backgroundwhen no absorption exists, the background signal is zero. Such dark background enables sensitive detection. In the presence of absorption, the foreground
signal is proportional to the absorption coefficient. Any small
fractional change in optical absorption coefficient translates
into an equal amount of fractional change in PAT signal,
which means a relative sensitivity of 100%.2 When the frac-
5760
5760
Prediction 2: Melanoma detection will become a high impact application of photoacoustic microscopy.
While melanoma is the most deadly form of skin cancer,
it is curable if detected early. The diagnosis of melanoma is
often based on its appearance. Abnormally appearing skin
lesions are resected for biopsy. Because visual inspection is
inaccurate and biopsy is invasive, an accurate noninvasive
method of melanoma detection is desirable.
PAM can acquire 1 structural images measuring tumor
burden and depth, 2 functional images of hemoglobin concentration measuring tumor angiogenesisa hallmark of
cancer, 3 functional images of hemoglobin oxygen saturation SO2 measuring tumor hypoxia or hypermetabolism
another hallmark of cancer, and 4 images of melanin concentration measuring tumor pigmentationa hallmark of
melanotic melanoma, consisting of 90% of melanomas
note that even amelanotic melanoma contains a low concentration of melanin. An in vivo PAM image of a melanoma in
a small animal is shown in Fig. 1. The depth of melanoma,
also an important prognostic parameter, was measured by
PAM. By contrast, other high-resolution optical microscopy
technologies cannot provide the required penetration. Because of biological variability in most biological problems, a
5761
5761
Penetration
Primary contrast
0.5 mm
0.5 mm
1 mm
Up to 30 mm, scalable
Scattering, fluorescence
Fluorescence
Scattering, polarization
Absorption
single biomarker is unlikely able to provide accurate diagnosis. However, the combination of four complementary measures can significantly enhance the likelihood of high accuracy. In addition, as Doppler photoacoustic microscopy
matures, blood flow measurement can add more diagnostic
information. Therefore, it is expected that the combined rich
contrasts will provide the sensitivity and specificity needed
for the early detection of melanoma.
III.C. Photoacoustic endoscopy
Prediction 4: Simultaneous functional and molecular photoacoustic tomography will provide unprecedented sensitivity and specificity to cancer diagnosis.
While functional imaging is based on endogenous contrast, molecular imaging is based on exogenous contrast
which essentially stains invisible biomarkers in vivo. The
former maps physiological activities at the tissue or organ
level, whereas the latter images biological and pathophysiological processes at the molecular level. Both can be accomplished quantitatively and noninvasivelywith the exMedical Physics, Vol. 35, No. 12, December 2008
5762
Prediction 5: Deep penetrating high-resolution photoacoustic tomography will become a complementary tool to
conventional optical imaging of reporter genes.
Understanding the nature of genes during in vivo development and pathogenesis is of significant interest. Such understanding can be enabled by gene expression imaging, a
form of molecular imaging based on reporter genes. A reporter gene is a segment of DNA fused to another gene of
interest. Unlike the contrast-carrying molecular imaging
probe presented in the preceding section, reporter genes must
express into protein products to provide contrast. The two
fused genes, sharing the same promoter, are transcribed into
a single mRNA molecule, which is then translated into fused
proteins. The protein encoded by the reporter gene carries
contrast either directly or through enzymatic reactions. As a
result, a positive contrast in a gene expression image indicates up-regulations of both the reporter gene and the fused
gene. Alternatively, a reporter gene can be used without gene
Medical Physics, Vol. 35, No. 12, December 2008
5762
Prediction 6: Doppler photoacoustic tomography will provide imaging of blood flow in vivo with high spatial and
velocity resolution.
The photoacoustic Doppler effect was recently discovered
experimentally.43,44 While the optical Doppler effect involves
light only, the acoustic Doppler effect involves sound only.
By contrast, the photoacoustic Doppler effect involves both
light and sound, resulting from the combination of the photoacoustic and Doppler effects. Here, the photoacoustic wave
undergoes a Doppler shift when the photoacoustic source
the part of the light absorbing medium generating the photoacoustic wavehas motion relative to the acoustic detector.
An important application is to image blood flow in vivo
by measuring the photoacoustic Doppler effect due to moving red blood cells. We coined photoacoustic Doppler flowmetry PADF for this imaging technology. PADF has potential advantages over conventional Doppler flowmetry. First,
PAT can image blood vessels at a depth of a few millimeters
with a spatial resolution of tens of micrometers, whereas
Doppler optical coherence tomography can penetrate only
one optical transport mean free path. Second, PADF tracks
light absorbing particles instead of scattering particles. Because red blood cells have much higher optical absorption
than the background outside blood vessels, PADF should
have low background noise and high sensitivity. By contrast,
acoustic flowmetry suffers from background scattering and
consequently has difficulty measuring slow flows. Third, the
Doppler shift in PADF is nearly independent of the excitation
light, enabling accurate measurement of both speed and direction of flow even at large depths inside an optically scattering medium. In comparison, laser Doppler flowmetry suffers from multiple light scattering, limiting the sensing depth
and obscuring the information of flow direction. Finally,
5763
PADF is free of speckle artifacts. Therefore, PADF is expected to break fresh ground for in vivo flow measurement
with high spatial resolution and high velocity sensitivity.
III.G. Photoacoustic tomography of metabolic rate of
oxygen
5763
For patients with breast cancer but clinically negative axillae, biopsy of the sentinel lymph node SLNthe first
draining nodesfor axillary staging has become a standard
clinical practice. In this conventional method, both optical
dye and radioactive Tc-99 colloids are injected into the breast
tumor. Both contrast agents are accumulated by SLNs. Emission from radioactivity is first detected to locate approximately the SLNs. Then, the SLNs are located precisely
through an invasive surgical procedure by visually tracing
the accumulated optical dye such as methylene blue. The
identified SLNs are then resected for pathological examination. Unfortunately, the surgical procedure has associated
morbidity. A noninvasive method for accurately locating the
SLN in vivo is sought-after because noninvasive diagnostic
methods such as fine needle biopsy could be subsequently
utilized without surgery.
PAT can potentially map the SLN noninvasively with high
spatial resolution in real time, avoiding invasive surgery and
harmful radioactivity. Preliminary data demonstrated that
PAT has both the required penetration depth and spatial resolution for SLN mapping.49 PAT can identify whether a node
is sentinel by detecting the accumulated dye because the dye
has high optical absorption contrast. The peak absorption
wavelength of methylene blue dye is 690 nm, which lies near
the optimal optical penetration window. Since the optical
contrast agent has been used in clinical SLN biopsy already,
avoiding FDA approval for a new optical contrast agent can
significantly accelerate the translation of PAT into the
clinic.50 While PAT has broad potential applications, imaging
the SLN is a perfect niche clinical application of PAT with
anticipated high impact on breast axillary staging and management.
III.I. Multiscale photoacoustic imaging in vivo
with common signal origins
5764
5764
Prediction 10: Simultaneous photoacoustic and thermoacoustic tomography of the breast will supplement or supplant
x-ray mammography.
While breast cancer remains the leading cause of cancer
death among women, early diagnosis improves survival.
X-ray mammography, the only mass screening tool for detecting nonpalpable breast cancers, has been shown to reduce
breast cancer deaths. However, ionizing radiation is required,
and detecting dense breasts and early-stage tumors is met
with limited sensitivity. In addition, imaging tumors close to
the chest wall is difficult. Therefore, alternative nonionizing
radiation-based screening technologies are needed. So far,
ultrasound imaging has been used as an adjunct to x-ray
mammography, especially in differentiating cystic from solid
breast masses. Magnetic resonance imaging has high sensitivity but variable specificity. It is further limited by its high
cost and use of exogenous contrast agent.
PAT and TAT can be integrated into conventional ultrasound imaging to provide multiple complementary contrasts
simultaneously. Three types of images of the breast can be
acquired: 1 structural ultrasound pulse-echo images, 2
functional laser-induced PAT images of concentration and
oxygen saturation of hemoglobin, and 3 radio frequencyinduced TAT images of water and sodium concentrations.
Good optical contrast, related to hallmarks of cancer, between tumor and normal breast tissues has been observed.53
In TAT, up to 5:1 contrast was observed between tumor and
normal breast tissues.29 In comparison, x-ray contrast among
soft tissues is typically only a few percent. Again, a single
contrast is usually unable to provide accurate diagnosis because of biological variability. However, the combination of
multiple complementary measures will likely improve the
accuracy.
While compatible with ultrasound imaging, PAT and TAT
are also compatible with each other, sharing the same ultrasonic detection system and image reconstruction algorithm.
Integrating PAT with TAT adds significant contrast but does
not add significant costs. PAT and TAT can potentially be
distributed to physicians as an add-on feature to existing
clinical ultrasound imaging systems. While PAT and TAT are
emerging technologies, ultrasonography is a mature clinical
technology. Incorporating PAT and TAT into conventional
ultrasonography will facilitate physicians acceptance of the
new technologies.
5765
Prediction 11: Photoacoustic and thermoacoustic tomography will provide low-cost, high-resolution, real-time, and
even bedside functional imaging of the brain.
Existing high-resolution human brain imaging
modalitiesincluding x-ray computed tomography, magnetic resonance imaging, and ultrasonographyhave limitations. Both x-ray computed tomography and magnetic resonance imaging are expensive and bulky. Further, x-ray
computed tomography uses ionizing radiation, and magnetic
resonance imaging requires a strong magnetic field. Therefore, it is advantageous to develop an affordable, nonionizing, high-resolution, and real-time imaging modality that can
be used in operating rooms or at the bedside. Such a system
can be used to monitor brain functions and conditions such
as strokes, head injuries, tumors, and brain infections. In certain cases, ultrasound brain imaging comes close to meeting
these objectives. Ultrasonography is an established pediatric
brain imaging modality when used before the fontanels are
closed. After the closure of the fontanels, the image quality
degrades significantly because the skull severely attenuates
and distorts ultrasonic waves.
Fortunately, unlike pulse-echo ultrasound imaging, PAT
and TAT involve only one-way ultrasound attenuation and
distortion due to the skull. As demonstrated previously,19 ultrasound penetration through small animal skulls is feasible,
and the associated distortions are relatively small. The authors group also demonstrated TAT through Rhesus monkey
skulls.54 The extension of PAT and TAT to human brain imaging is an area of research. The primary task is to correct
for the phase distortion due to the skull. The skull basically
functions as an irregular lens, whose aberration effect can be
compensated for if the phase distortion can be quantified. A
potential solution is to use geometric measurements of the
skull from x-ray computed tomography or magnetic resonance imaging to predict the phase distortion.
PAT and TAT can provide rich contrasts for highresolution structural and functional imaging. As demonstrated by diffuse optical tomography, near-infrared light can
penetrate through the skull to the cortex and has been used
for functional brain imaging at low spatial resolution. PAT
can potentially provide similar penetration but higher resolution. At an appropriate frequency, radio frequency can penetrate deeper than light. Therefore, TAT can potentially image deeper structures of the brain, while PAT will likely be
used to image cortical functions.
PAT and TAT are also relatively low-cost, real-time, and
portable. In comparison to x-ray computed tomography and
magnetic resonance imaging, ultrasonography is low cost.
Medical Physics, Vol. 35, No. 12, December 2008
5765
5766
5766
9
G. Ku and L. V. Wang, Scanning electromagnetic-induced thermoacoustic tomography: Signal, resolution, and contrast, Med. Phys. 28, 410
2001.
10
K. P. Kostli, D. Frauchiger, J. J. Niederhauser, G. Paltauf, H. P. Weber,
and M. Frenz, Optoacoustic imaging using a three-dimensional reconstruction algorithm, IEEE J. Sel. Top. Quantum Electron. 7, 918923
2001.
11
M. H. Xu, G. Ku, and L. V. Wang, Microwave-induced thermoacoustic
tomography using multi-sector scanning, Med. Phys. 28, 19581963
2001.
12
G. Paltauf, J. A. Viator, S. A. Prahl, and S. L. Jacques, Iterative reconstruction algorithm for optoacoustic imaging, J. Acoust. Soc. Am. 112,
15361544 2002.
13
M. H. Xu and L. V. Wang, Time-domain reconstruction for thermoacoustic tomography in a spherical geometry, IEEE Trans. Med. Imaging 21,
814822 2002.
14
Y. Xu, D. Z. Feng, and L. V. Wang, Exact frequency-domain reconstruction for thermoacoustic tomography-I: Planar geometry, IEEE Trans.
Med. Imaging 21, 823828 2002.
15
Y. Xu, M. H. Xu, and L. V. Wang, Exact frequency-domain reconstruction for thermoacoustic tomography-II: Cylindrical geometry, IEEE
Trans. Med. Imaging 21, 829833 2002.
16
V. G. Andreev, A. A. Karabutov, and A. A. Oraevsky, Detection of
ultrawide-band ultrasound pulses in optoacoustic tomography, IEEE
Trans. Ultrason. Ferroelectr. Freq. Control 50, 13831390 2003.
17
D. Finch, S. K. Patch, and Rakesh, Determining a function from its mean
values over a family of spheres, SIAM J. Math. Anal. 35, 12131240
2003.
18
K. P. Kostli and P. C. Beard, Two-dimensional photoacoustic imaging by
use of Fourier-transform image reconstruction and a detector with an
anisotropic response, Appl. Opt. 42, 18991908 2003.
19
X. D. Wang, Y. J. Pang, G. Ku, X. Y. Xie, G. Stoica, and L. V. Wang,
Noninvasive laser-induced photoacoustic tomography for structural and
functional in vivo imaging of the brain, Nat. Biotechnol. 21, 803806
2003.
20
X. D. Wang, Y. J. Pang, G. Ku, G. Stoica, and L. V. Wang, Threedimensional laser-induced photoacoustic tomography of mouse brain with
the skin and skull intact, Opt. Lett. 28, 17391741 2003.
21
M. H. Xu and L. V. Wang, Analytic explanation of spatial resolution
related to bandwidth and detector aperture size in thermoacoustic or photoacoustic reconstruction, Phys. Rev. E 67, 056605 2003.
22
M. Haltmeier, O. Scherzer, P. Burgholzer, and G. Paltauf, Thermoacoustic computed tomography with large planar receivers, Inverse Probl. 20,
16631673 2004.
23
G. Ku, X. D. Wang, G. Stoica, and L. V. Wang, Multiple-bandwidth
photoacoustic tomography, Phys. Med. Biol. 49, 13291338 2004.
24
Y. Xu and L. V. Wang, Time reversal and its application to tomography
with diffracting sources, Phys. Rev. Lett. 92, 033902 2004.
25
B. T. Cox and P. C. Beard, Fast calculation of pulsed photoacoustic fields
in fluids using k-space methods, J. Acoust. Soc. Am. 117, 36163627
2005.
26
G. Ku and L. V. Wang, Deeply penetrating photoacoustic tomography in
biological tissues enhanced with an optical contrast agent, Opt. Lett. 30,
507509 2005.
27
G. Ku, X. D. Wang, X. Y. Xie, G. Stoica, and L. V. Wang, Imaging of
tumor angiogenesis in rat brains in vivo by photoacoustic tomography,
Appl. Opt. 44, 770775 2005.
28
J. Zhang, M. A. Anastasio, X. C. Pan, and L. V. Wang, Weighted expectation maximization reconstruction algorithms for thermoacoustic tomography, IEEE Trans. Med. Imaging 24, 817820 2005.
29
G. Ku, B. D. Fornage, X. Jin, M. H. Xu, K. K. Hunt, and L. V. Wang,
Thermoacoustic and photoacoustic tomography of thick biological tissues toward breast imaging, Technol. Cancer Res. Treat. 4, 559565
2005.
30
L. Li, R. J. Zemp, G. Lungu, G. Stoica, and L. V. Wang, Photoacoustic
imaging of lacZ gene expression in vivo, J. Biomed. Opt. 12, 02050413 2007.
31
M. Li, J. Oh, X. Xie, G. Ku, W. Wang, C. Li, G. Lungu, G. Stoica, and L.
V. Wang, Simultaneous molecular and hypoxia imaging of brain tumors
in vivo using spectroscopic photoacoustic tomography, Proc. IEEE 96,
481489 2008.
32
L. V. Wang and G. Liang, Absorption distribution of an optical beam
focused into a turbid medium, Appl. Opt. 38, 49514958 1999.
5767
33
5767
46
S. H. Yee, K. Lee, P. A. Jerabek, and P. T. Fox, Quantitative measurement of oxygen metabolic rate in the rat brain using microPET imaging of
briefly inhaled O-15-labelled oxygen gas, Nucl. Med. Commun. 27,
573581 2006.
47
D. A. Boas, G. Strangman, J. P. Culver, R. D. Hoge, G. Jasdzewski, R. A.
Poldrack, B. R. Rosen, and J. B. Mandeville, Can the cerebral metabolic
rate of oxygen be estimated with near-infrared spectroscopy?, Phys.
Med. Biol. 48, 24052418 2003.
48
J. P. Culver, T. Durduran, T. Furuya, C. Cheung, J. H. Greenberg, and A.
G. Yodh, Diffuse optical tomography of cerebral blood flow, oxygenation, and metabolism in rat during focal ischemia, Biopolymers 23,
911924 2003.
49
K. H. Song, E. W. Stein, J. A. Margenthaler, and L. V. Wang, Noninvasive photoacoustic identification of sentinel lymph nodes containing methylene blue in vivo in a rat model, J. Biomed. Opt. 13, 054033-16
2008.
50
E. M. Sevick-Muraca, R. Sharma, J. C. Rasmussen, M. V. Marshall, J. A.
Wendt, H. Q. Pham, E. Bonefas, J. P. Houston, L. Sampath, K. E. Adams,
D. K. Blanchard, R. E. Fisher, S. B. Chiang, R. Elledge, and M. E.
Mawad, Imaging of lymph flow in breast cancer patients after microdose
administration of a near-infrared flurophore: Feasibility study, Radiology
246, 734741 2008.
51
K. Song and L. V. Wang, Deep reflection-mode photoacoustic imaging
of biological tissue, J. Biomed. Opt. 12, 060503 2007.
52
K. Maslov, H. F. Zhang, S. Hu, and L. V. Wang, Optical-resolution
photoacoustic microscopy for in vivo imaging of single capillaries, Opt.
Lett. 33, 929931 2008.
53
Q. Zhu, C. Xu, P. Y. Guo, A. Aquirre, B. H. Yuan, F. Huang, D. Castilo,
J. Gamelin, S. Tannenbaum, M. Kane, P. Hedge, and S. Kurtzman, Optimal probing of optical contrast of breast lesions of different size located
at different depths by US localization, Technol. Cancer Res. Treat. 5,
365380 2006.
54
Y. Xu and L. V. Wang, Rhesus monkey brain imaging through intact
skull with thermoacoustic tomography, IEEE Trans. Ultrason. Ferroelectr. Freq. Control 53, 542548 2006.
55
M. Pramanik, M. Swierczewska, D. Green, B. Sitharaman, and L. V.
Wang, Carbon nanotubes as a multimodalthermoacoustic and
photoacousticcontrast agent unpublished.
Received 6 August 2008; revised 5 December 2008; accepted for publication 8 December 2008;
published 26 January 2009
The first patients were treated with proton beams in 1955 at the Lawrence Berkeley Laboratory in
California. In 1970, proton beams began to be used in research facilities to treat cancer patients
using fractionated treatment regimens. It was not until 1990 that proton treatments were carried out
in hospital-based facilities using technology and techniques that were comparable to those for
modern photon therapy. Clinical data strongly support the conclusion that proton therapy is superior
to conventional radiation therapy in a number of disease sites. Treatment planning studies have
shown that proton dose distributions are superior to those for photons in a wide range of disease
sites indicating that additional clinical gains can be achieved if these treatment plans can be reliably
delivered to patients. Optimum proton dose distributions can be achieved with intensity modulated
protons IMPT, but very few patients have received this advanced form of treatment. It is anticipated widespread implementation of IMPT would provide additional improvements in clinical
outcomes. Advances in the last decade have led to an increased interest in proton therapy. Currently,
proton therapy is undergoing transitions that will move it into the mainstream of cancer treatment.
For example, proton therapy is now reimbursed, there has been rapid development in proton therapy
technology, and many new options are available for equipment, facility configuration, and financing. During the next decade, new developments will increase the efficiency and accuracy of proton
therapy and enhance our ability to verify treatment planning calculations and perform quality
assurance for proton therapy delivery. With the implementation of new multi-institution clinical
studies and the routine availability of IMPT, it may be possible, within the next decade, to quantify
the clinical gains obtained from optimized proton therapy. During this same period several new
proton therapy facilities will be built and the cost of proton therapy is expected to decrease, making
proton therapy routinely available to a larger population of cancer patients. 2009 American
Association of Physicists in Medicine. DOI: 10.1118/1.3058485
Key words: proton therapy, particle therapy, hadron therapy, radiation oncology
I. ABBREVIATED HISTORY OF PROTON THERAPY
E. Rutherford proposed the existence of protons in 1919;1
however, it was not until 1955 that the first patients were
treated with proton beams at the Lawrence Berkeley Laboratory LBL in California.2 In the years between 1919 and
1955, two events occurred that had an important impact on
proton therapy. First, in 1930, E. O. Lawrence built the first
cyclotron, paving the way for future particle accelerators
with energies high enough for cancer treatment applications.
In 1946, Robert Wilson then proposed that because of their
physical properties, beams of protons would be advantageous for the treatment of deep-seated cancers stating, It
will be easy to produce well collimated narrow beams of fast
protons, and since the range of the beam is easily controllable, precision exposure of well defined small volumes
within the body will soon be feasible.3 Wilson also described the use of a rotating wheel of variable thickness, i.e.,
a range modulation wheel RMW, interposed in a proton
beam as a method of adding together several Bragg peaks of
variable energies and weights in order to spread the proton
stopping region in depth to deliver a uniform dose to an
extended target volume. Figure 1 illustrates the concepts proposed by Wilson.
556
0094-2405/2009/362/556/13/$25.00
556
557
Photons
557
15 MV Photons
vs.
Protons
Ideal dose
distribution
Protons
each directed from a different angle, this high dose pull back
into normal tissue is not additive over all fields. In principle,
the use of an RMW to obtain an SOBP in passive scattering
techniques modulates both energy and beam intensity. However, this should not be confused with intensity modulated
proton treatment IMPT achieved with beam scanning techniques as described below.
The second type of beam delivery utilizes spot scanning
techniques also often called pencil beam scanning. Figure 3
illustrates how a target can be scanned by placing Bragg
peaks throughout the volume by the use of scanning magnets
and energy changes. Energy changes in scanning techniques
can be carried out with various methods including: 1 energy changes in the accelerator when a synchrotron is used;
2 energy changes made with an energy selection system
when a cyclotron is used; or 3 either of the above methods
plus energy absorbers in the treatment nozzle.
558
558
High-density
structure
Scanning Magnets
Critical
Structure
Scanning Magnets
Proton Pencil
Beam
FIG. 3. Diagram of a typical pencil beam scanning system. Two sets of scanning magnets scan the beam in a
2D pattern: the beam range is adjusted by changing the
beam energy entering the nozzle. In the usual case, all
spots for the deepest range are scanned, the energy is
changed, and all spots with the new range are scanned,
etc., until the entire target volume has been scanned. In
pencil beam scanning, the high dose region can be confined to the target volume and no high dose spills over
into normal tissue.
Body
Surface
Target
Volume
559
Between the mid-1980s and mid-1990s, medical physicists developed methods to fully utilize medical imaging and
to calculate three-dimensional 3D treatment plans for external beams of photons, electrons, protons, and light ions. In
addition, inverse planning and optimization techniques were
developed that made it possible to calculate treatment plans
for intensity modulated radiation therapy. These developments led to a large number of treatment planning comparisons including those for:
Medical Physics, Vol. 36, No. 2, February 2009
559
560
Until 1990, all proton therapy took place in researchbased facilities with equipment developed by the clinical researchers and engineers who worked in those facilities. The
lack of a sizable market was a disincentive for manufacturers
to spend large sums of money to develop proton therapy
equipment. The confluence of factors during the 1990s and
Medical Physics, Vol. 36, No. 2, February 2009
560
early 2000s discussed above created a more favorable climate for vendors and there are currently several companies
marketing proton therapy systems and offering systems that
provide both protons and carbon ions for cancer therapy.
New proton/carbon systems based on superconducting technology have been developed that will make it possible to
build more compact isocentric gantries and accelerators for
hospital-based systems. In addition, companies have been
formed to develop and operate proton therapy facilities and
others offer various levels of consulting e.g., feasibility
studies, marketing studies, business plans, technology evaluation, staffing and operating plans, etc.. The success of new
hospital-based proton therapy projects like those at the University of Florida/Shands Cancer Center in Jacksonville,
Florida and The University of Texas M. D. Anderson Cancer
Center in Houston, Texas has demonstrated that these complex, expensive facilities can be built on time and on budget,
and can rapidly reach targeted patient treatment capacity in
order to satisfy ambitious clinical programs and business
plans.
III.F. Summary
MGH and LLUMC have carried out dose escalation studies for prostate cancer.24 Recently, the National Cancer Institute approved funding for a P01 grant application submitted
by MGH and M. D. Anderson Cancer Center MCACC to
conduct clinical studies in proton therapy with the objective
of applying advanced proton radiation planning and delivery
techniques to improve outcomes for patients with nonsmallcell lung cancer proton dose escalation and proton vs. photon randomized trials, liver tumors, pediatric medulloblastoma and rhabdomyosarcoma, spine/skull base sarcomas,
and paranasal sinus malignancies. It is expected that the University of Florida and University of Pennsylvania facilities
will also be participating in multi-institutional clinical
studies.
561
561
cheaper per treatment room as compared to large systems using single accelerators to feed several treatment
rooms.
Additional rooms may have to be installed if demand
for proton therapy increases in a location with a singleroom system. If several treatment rooms are eventually
needed, the cost effectiveness of the single-room concept may be lost; for example, an additional accelerator
will be needed for each treatment room. Maintaining
multiple accelerators will also increase maintenance
costs.
Because of the pulse structure of some accelerators
e.g., synchrocyclotrons chosen for the proposed systems, it may be difficult to use spot scanning techniques
to deliver IMPT treatments.28 IMPT could be delivered
using a multileaf collimator; however, this method may
have problems such as poor efficiency in the use of
protons and increased neutron production.
These systems are new, and, thus, their operability, reliability, and maintainability are not known.
IV.C.2. Accelerator development
Advances in accelerator technology are also expected.
Several developments already underway show considerable
potential to provide a new generation of more compact, more
efficient, and less expensive accelerators for proton therapy.
Some developments will improve upon current technologies,
while others will provide entirely new types of accelerators,
some of which are highlighted here. Flanz29 has provided an
excellent overview of new accelerator technologies.
IV.C.2.a. Superconducting cyclotrons and synchrocyclotrons. Cyclotron and synchrocyclotron development has resulted in more compact systems with increased energy. These
gains have been achieved by the use of new accelerator designs and the application of superconducting technology.
Figure 5 illustrates how the weight of cyclotrons/
synchrocyclotrons has deceased since 1995. Notably, the accelerator energy has increased while the weight/size of the
accelerators has decreased. At the current reduced weights of
approximately 20 tons, accelerators can be installed on isocentric gantries and rotated around patients, eliminating large
and expensive beam transport systems.19,30 There is at least
one vendor marketing a one-room proton therapy system
based on this concept. Such systems will be particularly attractive to small community hospitals and large radiation oncology private practices.
IV.C.2.b. Fixed field alternating gradient (FFAG) accel
erators. To date, most particle therapy facilities have used
isochronous cyclotrons, and synchrotrons for particle acceleration. However, FFAG accelerators may be able to replace
both cyclotrons and synchrotrons for hadron therapy. FFAG
accelerators combine many of the positive features of cyclotrons and synchrotrons with fixed magnetic fields as in cyclotrons and pulsed acceleration as in synchrotrons. FFAG
accelerators have the following potential advantages:
FFAG accelerators can be cycled faster than synchrotrons, limited only by the rate of the radiofrequency
Medical Physics, Vol. 36, No. 2, February 2009
562
250
IBA 230
200
150
Tons
562
100
ACCEL K250
50
New
0
1995
IBA 230
1999
2003
ACCEL
K250
2006
New
2010
Date
FIG. 5. This graph indicates how the weight and, therefore, the size of
clinical cyclotrons has decreased since 1995. The IBA 230 room temperature cyclotron was the first cyclotron to be installed in a hospital-based
proton therapy facility. More recently, the ACCEL K250 now the Varian/
ACCEL K250 has been installed in two European facilities. New superconducting cyclotrons will weigh about 10% of the IBA 230.
563
563
564
564
also been quite useful in calculating data used in commissioning proton treatment planning systems47,48 MC calculations can decrease the time required to commission a treatment planning system by eliminating a large number of
measurements, especially in the case of spot beam scanning
where approximately 100 energies need to be commissioned.
It is reasonable to expect that, with future developments,
particularly in the area of increasing the speed of MC calculations, MC will play an increasing role in proton treatment
planning.
IV.C.4.b. Optimization for IMPT treatment planning. Protons are much more sensitive to tissue inhomogeneities in the
beam path than photons and current optimization algorithms
do not accurately account for the errors resulting from range
uncertainties and changes in scattering conditions caused by
such inhomogeneities. For passive-scattering techniques,
such uncertainties are partially offset by expanding the margins around the clinical target volume or by using internal
target volumes determined from 4D CT treatment planning
scans. In IMPT, the dose distribution is sensitive to the uncertainties in each individual beam of the potentially several
thousand Bragg peaks used in the treatment plan, and uncertainties in the spot placements can result in unacceptable hot
and cold regions in the target volume. One approach to improving IMPT treatment planning is to include errors in the
optimization calculations in such a way as to ensure that the
treatment plan predicts actual dose distributions more reliably. Work in this area is underway and these approaches
have the potential to improve the accuracy and robustness of
IMPT treatment plans.49
IV.C.5. Positron emission tomography PET
for treatment verification
Ideally, one would have a method available to measure
proton dose distributions delivered to patients either online
during treatment or offline immediately or soon after treatment. Such treatment verification would make it possible to
correct the treatment plan after the first treatment session or
to make corrections during the course of treatment. Such
adjustments may be necessitated by changes in the patients
weight or changes in the tumors size, shape, or position
during treatment. Positron emission tomography PET is a
potentially useful tool for validating the fidelity of the treatment plan delivery.
PET can verify treatment delivery by detecting annihilation -rays arising from the decay of small amounts of +
emitters such as 11C, 15O, and 10C produced by nuclear fragmentation reactions between the primary charged particle
and the target nuclei in the irradiated tissue. Particle treatment delivery verification can be achieved by comparing the
measured + activity distribution with an expected pattern
calculated by treatment planning or by MC calculations of
the expected + activity. In particular, PET techniques offer
the potential to detect and quantify range uncertainties in
treatment planning calculations.
Physicists at MGH and elsewhere5053 have been exploring the use of PET imaging, both offline and online, for
565
treatment verification. The preliminary results look promising and it is expected that with further development, this
technique will become an important tool for verifying particle treatment plan delivery.
IV.C.6. Proton CT
The idea of using proton beams for imaging has been
around for some time. In his seminal papers on the application of photon absorption in radiology, Cormack pointed out
the possibility of using heavy charged particles in CT
applications.54,55 In 1968, Koehler demonstrated that
160 MeV protons could produce images on radiographic
films that had much better contrast than those produced with
photons.56 Cormack and Koehler published a study on proton
CT in 1976.57 In the late 1970s and early 1980s, Hanson et
al. published a series of papers on proton CT including one
of the earliest studies of human specimens.58 In addition,
Schneider et al. conducted studies on the use of proton radiography as a quality assurance tool for proton treatment
planning.59,60 These studies, including measurements taken
on an animal patient, concluded that inaccuracies in treatment planning as a result of incomplete modeling of multiple
Coulomb scattering effects and errors in proton stopping
powers derived from photon CT scans could result in sizeable deviations in predicted versus measured proton dose
distributions. Zygmanski et al. demonstrated the feasibility
of using proton cone-beam CT to determine proton stopping
powers for materials of various densities.61 In 2004, Schulte
et al. at the LLUMC proton therapy center reported on a
conceptual design for a proton CT system for applications in
proton radiation therapy.62
Currently, most proton treatment planning dose calculations use a pencil beam algorithm that accounts for tissue
inhomogeneities by means of pathlength water equivalent
depth scaling based on relative stopping powers. Hounsfield
numbers obtained from x-ray treatment planning CT scans
are converted to proton stopping powers using measured relationships between Hounsfield numbers and the relative
stopping powers for materials closely matched to human tissues such as soft tissue, fat, lung tissue, soft bone, and hard
bone.63 These conversions are frequently inexact, which can
lead to errors in the calculation of proton ranges in patients
of approximately 3% or more of the proton range. These
errors could be substantially reduced by the direct measurement of relative proton stopping powers using proton CT
techniques.
There are, however, also errors in proton CT data. For
instance, limitations in the spatial and density resolution of
proton CT arise from various sources: multiple Coulomb
scattering in the patient leads to spatial uncertainties in the
CT image and energy loss straggling due to momentum
spread of the beam entering the patient and proton range
straggling in the patient can cause errors in density resolution. In addition, secondary protons arising from nonelastic
nuclear interactions can lead to noise in the proton CT images. Recent studies have demonstrated that density resolutions of 1%2% are possible with proton CT techniques,
Medical Physics, Vol. 36, No. 2, February 2009
565
566
566
V. CONCLUSIONS
Proton therapy is in the midst of a remarkable transition.
During the next decade, important advances in proton
therapy treatment delivery, treatment planning, and quality
assurance systems will be made that will improve proton
therapys efficiency, robustness, and accuracy. It is also expected that the cost to patients will decrease, making proton
therapy more financially competitive with photon therapy.
IMPT treatments will become available for routine use in
an increasing number of facilities, and methods will be developed to improve the accuracy and precision of this treatment modality. With the availability of advanced IMPT techniques, it may be possible to quantify the clinical gains
obtained from optimized proton therapy and to compare the
clinical outcomes of photons and protons using the best dose
distributions for both modalities.
It will be necessary to develop methods to make proton
treatment plan calculations more accurate and to verify that
treatment plans are actually realized in the delivery of proton
treatments. It is not meaningful to compare protons with
photons unless we can demonstrate that the superior dose
distributions of proton treatment plans can actually be delivered to patients.78
More hospital-based, state-of-the-art proton therapy facilities will be built and increasing numbers of patients will be
treated. As stated by Schultz-Ertner and Tsujii, The potential of particle therapy can only be exploited if a full integration of particle therapy into clinical environments and interdisciplinary treatment strategies is sought and if new medical
and technologic advances are properly incorporated into the
total treatment process.15
a
567
12
567
37
568
568
70
D. Nichiporov and K. Solberg, Multichannel detectors for profile measurements in clinical proton fields, Med. Phys. 34, 26832690 2007.
71
S. Boon, P. van Luijk, T. Bhringer, A. Coray, A. Lomax, E. Pedroni, B.
Schaffner, and J. M. Schippers, Performance of a fluorescent screen and
CCD camera as a two dimensional system for dynamic treatment techniques, Med. Phys. 27, 21982208 2000.
72
S. Vatnitsky, Radiochromic film dosimetry for clinical proton beams,
Appl. Radiat. Isot. 48, 643651 1997.
73
D. Nichiporov et al., Investigation of applicability of alanine and radiochromic detectors to dosimetry of proton clinical beams, Appl. Radiat.
Isot. 46, 13551362 1995.
74
W. B. Warren, Evaluation of Bang polymer gel dosimeters in proton
beams, M.S. thesis, The University of Texas Health Science Center at
Houston, Graduate School of Biomedical Sciences, 2007.
75
G. Kraft, The radiobiological and physical basis for radiotherapy with
protons and heavier ions, Strahlenther. Onkol. 166, 1013 1990.
76
T. Kanai et al., Biophysical characteristics of HIMAC clinical irradiation
system for heavy-ion radiation therapy, Int. J. Radiat. Oncol., Biol.,
Phys. 64, 201210 1999.
77
A. Brahme, Recent advances in light ion radiation therapy, Int. J. Radiat. Oncol., Biol., Phys. 58, 603616 2004.
78
R. Mohan, Achieving what-you-see-is-what-you-get in proton therapy,
PTCOG Scientific Meeting, Jacksonville, Fl, May 2008, http://
ptcog.web.psi.ch/ptcog47_talks.html accessed November 1, 2008.
Received 23 July 2008; revised 2 February 2009; accepted for publication 27 March 2009;
published 5 May 2009
Tomosynthesis is a decades-old technique for section imaging that has seen a recent upsurge in
interest due to its promise to provide three-dimensional information at lower dose and potentially
lower cost than CT in certain clinical imaging situations. This renewed interest in tomosynthesis
began in the late 1990s as a new generation of flat-panel detectors became available; these detectors
were the one missing piece of the picture that had kept tomosynthesis from enjoying significant
utilization earlier. In the past decade, tomosynthesis imaging has been investigated in a variety of
clinical imaging situations, but the two most prominent have been in breast and chest imaging.
Tomosynthesis has the potential to substantially change the way in which breast cancer and pulmonary nodules are detected and managed. Commercial tomosynthesis devices are now available or
on the horizon. Many of the remaining research activities with tomosynthesis will be translational
in nature and will involve physicist and clinician alike. This overview article provides a forwardlooking assessment of the translational questions facing tomosynthesis imaging and anticipates
some of the likely research and clinical activities in the next five years. 2009 American Association of Physicists in Medicine. DOI: 10.1118/1.3120285
Key words: tomosynthesis, breast imaging, chest imaging, tomography
I. OVERVIEW
Digital tomosynthesis is a simple and relatively inexpensive
method of producing section images using conventional digital x-ray equipment. It is a form of limited angle tomography
that produces section, or slice, images from a series of
projection images acquired as the x-ray tube moves over a
prescribed path. The total angular range of movement is often less than 40. Because the projection images are not acquired over a full 360 rotation about the patient, the resolution in the z direction i.e., in the depth direction
perpendicular to the x-y plane of the projection images is
limited, and thus tomosynthesis does not produce the isotropic spatial resolution achievable with computed tomography
CT. However, the resolution of images in the x-y plane of
the reconstructed slices is often superior to CT, and the ease
of use in conjunction with conventional radiography makes
tomosynthesis a potentially quite useful imaging modality.
There has been a high degree of research interest in tomosynthesis imaging in the past decade, and at least two
commercial products have recently been approved by the
Food and Drug Administration FDA and released on the
market. It is expected that other approved devices will soon
follow. As such, tomosynthesis imaging is in a period of a
high rate of change, with an increasing number of investigators and manufacturers nearing completion of projects involving both the physics and clinical aspects of the technique. If one were to compare the current state of
tomosynthesis imaging to a metaphorical calendar year, the
field is firmly in the middle of spring. It has moved beyond
the winter of initial research investigation but is not yet in
the summer as a mature and accepted clinical modality.
Just as the rate of change in daylight hours is at its greatest at
1956
the equinoxes and slows down near the solstices, so the rate
of progress in a new imaging modality is greatest in the
middle of its developmental pathway. Tomosynthesis imaging is clearly in March of its allegorical calendar year of
development. Change is happening at a rapid pace, which is
exciting for investigators, but it also makes the long-term
future difficult to predict with certainty.
Although tomosynthesis has not yet passed its pivotal
clinical evaluation, it can, to borrow another metaphor, be
said to have passed the light box test. A clinical colleague
has often remarked that he can gauge which new imaging
modalities will be clinically successful from an intuitive appraisal of how substantially the new technique appears to
improve upon the modality it is vying to replace by viewing
the images on a light box across the room. In the opinion
of this colleague, if improvements from a new modality are
only demonstrable by comparing small changes in Az values
following an extensive ROC study, then the improvement of
the new modality may be statistically significant but the level
of its clinical impact may be hard to predict in advance. On
the other hand, if the new modality produces images that are
so substantially superior to the predecessor technique that the
difference is visible on a light box viewed across the room,
then the clinical impact is likely to be substantial. After having viewed several thousand tomosynthesis images in early
clinical studies, most observers would agree that there is a
subjective but substantial improvement in the ability to appreciate abnormal anatomy or disease in tomosynthesis images relative to conventional radiography. At the current time
there are only a limited number of completed clinical studies
to quantify this improvement and to determine how tomosynthesis affects specificity as well as sensitivity. Nonethe-
0094-2405/2009/366/1956/12/$25.00
1956
1957
1957
1958
1958
FIG. 1. Images of pulmonary nodules in chest tomosynthesis images of a human subject. a Coned view of digital PA radiograph shows one clearly visible
right lung nodule arrow. b Tomosynthesis image shows the same nodule vertical arrow as seen on the PA radiograph in a. A second nodule horizontal
arrow is also visible that was not seen in the PA radiograph in a. c Tomosynthesis image at a more posterior level shows an additional left lung nodule
arrow not seen in the PA radiograph in a. d CT image lung window confirms left lower lobe nodule seen in c. Reprinted with permission from
Medical Physics, Ref. 16, Copyright 2008, American Association of Physicists in Medicine AAPM.
1959
1959
The most frequently used reconstruction algorithm for tomosynthesis is commonly referred to as shift and add SAA.
Medical Physics, Vol. 36, No. 6, June 2009
1960
1960
The optimum image acquisition parameters have been investigated in several laboratories and depend on both the
clinical application as well as the reconstruction algorithm
used. In the case of chest imaging, research in our own laboratory has determined that 71 projection images an odd
number is preferable acquired over 20 of tube movement is
best for detection of pulmonary nodules with the MITS
algorithm.15,16 A commercial device currently on the market
uses about 60 images acquired over 40 for chest imaging.
While the number of projection images is fairly consistent
between these two implementations, the angle obviously dif-
1961
fers substantially. The MITS algorithm used in our laboratory has an impulse response that performs better at
midrange angles15 and FBP used with the commercial devices performs better at wider angles,32 thus explaining this
difference. In application to breast imaging, anywhere from
11 to 49 projection images have been used,33,34 with a total
angle of tube movement in the range of 1550.33,35,36
The spacing of the reconstructed planes also varies with
the clinical application and the reconstruction algorithm. In
breast imaging, it is common to use 1 mm plane spacing,19,33
but in chest imaging, 5 mm is more typical.16
III.D. Commercial implementation
Commercial manufacturers have recently introduced tomosynthesis products that can be used for chest, abdominal,
and musculoskeletal applications. Two such devices are currently on the market and are FDA approved. Devices for
breast tomosynthesis are being developed by several manufacturers, and at least one manufacturer has a submission to
the FDA for breast imaging applications.37 It is likely that
within 1 year there will be an FDA-approved device on the
market for breast imaging in the U.S.
IV. TOWARD CLINICAL TRANSLATION
Tomosynthesis research over the past two decades has
addressed many of the fundamental physics questions, and
the field has reached a level of maturity reminiscent of the
first or second generation CT or MR scanners. With the introduction of commercial products, many more investigators
and clinicians will have access to tomosynthesis imaging,
and a host of new questions will arise regarding its appropriate clinical utilization. Indeed, an increased interest in tomosynthesis has already been seen, as reflected in the exponentially expanding number of papers and presentations on this
topic at recent scientific meetings. Many of the questions
ahead will largely be translational in nature, which is not to
say that physicists will not be actively involved; rather, research and applications in tomosynthesis will increasingly
involve the combined efforts of both physicists and clinicians. In this section, some of the likely translational activities will be explored, and a synthesis of where the field is
likely to go next will be given.
IV.A. Continued physics optimization
1961
1962
1962
large clinical trials. It will also be necessary to evaluate recall rate and biopsy rate, as well as whether the positive
predictive value for biopsy recommendation improves with
tomosynthesis.36 The higher sensitivity of tomosynthesis
may mean that more objects are seen that look suspicious,
and thus the number of recalls may initially increase; however, there is some evidence that tomosynthesis may allow a
better look at potential pathologies, thereby ruling out some
false positives, and thus ultimately the number of cases that
would be recalled may be reduced compared with conventional mammography.33 The very important translational
question of how tomosynthesis affects diagnostic decision
making and patient outcomes will need larger clinical studies
to resolve.47,51
A second important question is how many views to take
with breast tomosynthesis. Currently, some investigators feel
that tomosynthesis should be performed in both mediolateral
oblique MLO and craniocaudal CC views,52 although that
opinion is not universally held. There also remains a question
about whether or not a static view should also be obtained.
The answer to this question depends largely on how effective
tomosynthesis is at imaging microcalcifications. There are
several challenges with imaging microcalcifications with tomosynthesis, including the reduced resolution of the method
compared with conventional single-image mammography
and also the fact that the pattern of distribution of microcalcifications is more difficult to appreciate in section imaging
unless a relatively thick section is used. The reduced resolution with tomosynthesis relative to a single-view FFDM
image is due to the need to shift images by fractional pixel
amounts prior to adding during reconstruction. This reduced
resolution may impact the ability to appreciate the morphology of small calcifications, the importance of which is open
to debate among clinical observers. If it is decided that a
static view is needed to adequately evaluate calcifications,
then a scenario may develop where an MLO static view
mammogram is acquired along with MLO and CC view tomosynthesis exams. Nishikawa et al.53 have proposed a hybrid scheme where a regular MLO or CC view digital mammogram at normal dose is evaluated for microcalcifications,
and a reduced dose and reduced resolution tomosynthesis
data set is reviewed for masses. Clinical trials will be needed
to determine which of these is the most appropriate utilization scheme.
A third translational question for breast tomosynthesis is
how tomosynthesis used for screening will impact diagnostic
workup procedures. Tomosynthesis used for screening will
likely result in fewer diagnostic workups, and traditional
methods such as ultrasound and magnification view mammography can still be used to evaluate suspicious opacities
noted on screening tomosynthesis. Will tomosynthesis have
sufficient specificity that certain patients can go directly to
biopsy? And how will the very small number of cancers
e.g., circumscribed that do not display well on tomosynthesis be handled in screening? These practical questions of
utilization will likely be resolved as radiologists gain clinical
experience with tomosynthesis in the first few years after
FDA-approved devices are available.
1963
There are other new breast imaging approaches under investigation that may impact how tomosynthesis is used. One
example is the development of novel x-ray sources using
carbon nanotube field-emission cathodes.54 If these devices
are successful and reasonably priced, they may enable very
rapid collection of tomosynthesis projection images that
would eliminate the need for lengthy compression. A second
example is the use of iodinated contrast in breast imaging. If
injection of contrast is found useful in breast cancer imaging,
will it also be advantageous in tomosynthesis imaging? Initial studies indicate that it may be.55 If contrast-enhanced
tomosynthesis is found to be advantageous, it may be less
costly and more readily available than alternatives such as
contrast-enhanced magnetic resonance imaging. A third and
larger question is how breast tomosynthesis and cone-beam
CT of the breast will compare. CT is advantageous in that it
has better depth resolution than tomosynthesis, but it also
requires a larger room with more specialized equipment, requires the patient to lie prone, and has difficulty imaging
near the chest wall.56 Computer simulation studies indicated
that both tomosynthesis and breast CT outperformed digital
mammography for detection of masses, but tomosynthesis
and CT performed roughly comparably to each other.46 The
jury is out on this comparison, and studies comparing these
two modalities will be necessary to decide ultimately
whether CT or tomosynthesis will prove superior.
IV.C. Chest imaging
1963
1964
1964
Computer-aided detection CAD and computer-aided detection diagnosis CADx schemes are often mentioned in
connection with tomosynthesis73 and for two primary reasons. First, the reduction of overlying anatomy offers the
potential for improved sensitivity and specificity for CAD
algorithms. One of the factors that limits the success of some
CAD algorithms is the large number of false-positive findings. Using tomosynthesis to eliminate much of the confusing background should theoretically reduce the number of
false positives. However, in some cases such as with chest
tomosynthesis, this reduction in false positives from reduction of overlying soft-tissue might be limited by increased
false positives from circular cross sections of imaged vessels.
Thus, exactly how much tomosynthesis will benefit CAD
algorithms remains to be seen. There is ongoing research in
several laboratories, including ours, into these issues.
1965
1965
sensitivity and specificity of tomosynthesis in cancer imaging. Further studies are needed to address its full clinical
performance.
Tomosynthesis is in somewhat of a chicken and egg situation at the present time with regard to its clinical acceptance. Before tomosynthesis will be widely adopted in clinical use, its benefit to patient outcomes must be demonstrated
with further studies and wider clinical experience. However,
in order to complete such studies and gain such wider clinical experience, the technique must be used on more patients.
This type of impasse is often seen early in the adoption of
any new modality, and it is certain that gaining reimbursement for the technique will be an important component of
accelerating its clinical utilization.
Tomosynthesis is fundamentally at a translational crossroads at the present time, with many of the remaining questions not ones of physics optimization or even clinical performance but rather pertaining to practical matters. How will
tomosynthesis impact workflow, and what will be the consequences of that effect? How will tomosynthesis fit into the
paradigm of clinical management as a tool halfway between
traditional radiography and CT? Will patients that receive
tomosynthesis be sent on to CT or other standard diagnostic
workup afterwards in most cases anyway, thus ultimately
not saving much money for the healthcare enterprise? Or will
tomosynthesis find a niche that enables better utilization of
dose and imaging dollars than the existing clinical paradigm?
This author believes the latter will ultimately be the case,
although further trials are needed to demonstrate it. Tomosynthesis is somewhat unique as a new imaging modality in
that it is not likely to produce a revolution as CT did but
rather an evolution of better clinical management. We are
currently in the exciting and uncertain times when the tipping point will likely be seen in the next 13 years that will
determine the future of this interesting imaging modality.
ACKNOWLEDGMENTS
The author gratefully acknowledges the input and suggestions of Joseph Y. Lo, Ph.D., Jay A. Baker, MD, Devon J.
Godfrey, Ph.D., Michael J. Flynn, Ph.D., and Donald A. Tyndall, Ph.D., DDS in the preparation of this manuscript. Some
of the research results from the authors laboratory were supported by grants from NIH Grant No. R01 CA80490 and
GE Healthcare. GE and Duke jointly hold a patent related to
certain aspects of tube movement in tomosynthesis imaging.
Collaborative work on breast tomosynthesis at the authors
institution was supported in part by a grant from Siemens
Medical Solutions.
a
1966
1966
27
Y. Zhang, H. P. Chan, B. Sahiner, J. Wei, M. M. Goodsitt, L. M. Hadjiiski, J. Ge, and C. Zhou, A comparative study of limited-angle conebeam reconstruction methods for breast tomosynthesis, Med. Phys. 33,
37813795 2006.
28
Y. Zhang, H. P. Chan, B. Sahiner, Y.-T. Wu, C. Zhou, J. Ge, J. Wei, and
L. M. Hadjiiski, Application of boundary detection information in breast
tomosynthesis reconstruction, Med. Phys. 34, 36033613 2007.
29
R. Fahrig, A. R. Pineda, E. G. Solomon, A. N. Leung, and N. J. Pelc,
Fast tomosynthesis for lung cancer detection using the SBDX geometry, Proc. SPIE 5030, 371378 2003.
30
A. R. Pineda, S. Yoon, D. S. Paik, and R. Fahrig, Optimization of a
tomosynthesis system for the detection of lung nodules, Med. Phys. 33,
13721379 2006.
31
E. G. Solomon, M. S. Van Lysel, R. E. Melen, J. W. Moorman, and B.
Skillicorn, Low-exposure scanning-beam x-ray fluoroscopy system,
Proc. SPIE 2708, 140149 1996.
32
B. Li, G. B. Avinash, J. W. Eberhard, and B. E. H. Claus, Optimization
of slice sensitivity profile for radiographic tomosynthesis, Med. Phys.
34, 29072916 2007.
33
S. P. Poplack, T. D. Tosteson, C. A. Kogel, and H. M. Nagy, Digital
breast tomosynthesis: Initial experience in 98 women with abnormal digital screening mammography, AJR Am. J. Roentgenol. 189, 616623
2007.
34
W. Zhao, B. Zhao, P. R. Fisher, P. Warmoes, T. Mertelmeier, and J.
Orman, Optimization of detector operation and imaging geometry for
breast tomosynthesis, Proc. SPIE 6510, M1M12 2007.
35
T. Wu, R. H. Moore, and D. B. Kopans, Voting strategy for artifact
reduction in digital breast tomosynthesis, Med. Phys. 33, 24612471
2006.
36
J. M. Park, E. A. J. Franken, M. Garg, L. L. Fajardo, and L. T. Niklason,
Breast tomosynthesis: Present considerations and future applications,
Radiographics 27, S231S240 2007.
37
T. Wallack, New dimension in detection, Boston Globe, June 6, 2008.
38
Y. Chen, J. Y. Lo, J. A. Baker, and J. T. Dobbins III, Gaussian frequency
blending algorithm with matrix inversion tomosynthesis MITS and filtered back projection FBP for better digital breast tomosynthesis reconstruction, Proc. SPIE 6142, 0E10E9 2006.
39
Y. Chen, J. Y. Lo, and J. T. Dobbins III, Impulse response analysis for
several digital tomosynthesis mammography reconstruction algorithms,
Proc. SPIE 5745, 541549 2005.
40
J. T. Rakowski and M. J. Dennis, A comparison of reconstruction algorithms for C-arm mammography tomosynthesis, Med. Phys. 33, 3018
3032 2006.
41
J. Zhou, B. Zhao, and W. Zhao, A computer simulation platform for the
optimization of a breast tomosynthesis system, Med. Phys. 34, 1098
1109 2007.
42
H. C. Gifford, C. S. Didier, M. Das, and S. J. Glick, Optimizing breasttomosynthesis acquisition parameters with scanning model observers,
Proc. SPIE 6917, 0S10S9 2008.
43
T. Deller, K. N. Jabri, J. M. Sabol, X. Ni, G. Avinash, R. Saunders, and R.
UppaluriEffect of acquisition parameters on image quality in digital tomosynthesis, Proc. SPIE 6510, 1L11L11 2007.
44
I. Reiser, B. A. Lau, and R. M. Nishikawa, Effect of scan angle and
reconstruction algorithm on model observer performance in tomosynthesis, in IWDM 2008, edited by E. A. Krupinski Springer-Verlag, Berlin,
2008, Vol. LNCS 5116, pp. 606611.
45
H. P. Chan, J. Wei, Y. Zhang, B. Sahiner, L. M. Hadjiiski, and M. A.
Helvie, Detection of massses in digital breast tomosynthesis mammography: Effects of the number of projection views and dose, in IWDM
2008, edited by E. A. Krupinski Springer-Verlag, Berlin, 2008, Vol.
LNSC 5116, pp. 279285.
46
X. Gong, S. J. Glick, B. Liu, A. A. Vedula, and S. Thacker, A computer
simulation study comparing lesion detection accuracy with digital mammography, breast tomosynthesis, and cone-beam CT breast imaging,
Med. Phys. 33, 10411052 2006.
47
W. F. Good, G. S. Abrams, V. J. Catullo, D. M. Chough, M. A. Ganott, C.
M. Hakim, and D. Gur, Digital breast tomosynthesis: A pilot observer
study, AJR Am. J. Roentgenol. 190, 865869 2008.
48
J. Vikgren, S. Zachrisson, A. Svalkvist, . A. Johnsson, M. Boijsen, A.
Flinck, S. Kheddache, and M. Bth, Comparison of chest tomosynthesis
and chest radiography for detection of pulmonary nodules: Human observer study of clinical cases, Radiology 249, 10341041 2008.
49
C. M. Li and J. T. Dobbins III, Methodology for determining dose re-
1967
1967
62
Jack L. M. Venselaar
Department of Medical Physics, Instituut Verbeeten, P.O. Box 90120, 5000 LA Tilburg, The Netherlands
Luc Beaulieu
Dpartement de Radio-Oncologie et Centre de Recherche en Cancrologie de lUniversit Laval,
Centre Hospitalier Universitaire de Qubec, 11 Cte du Palais, Qubec, Qubec G1R 2J6, Canada
and Dpartement de Physique, de Gnie Physique et dOptique, Universit Laval, Qubec,
Qubec G1K 7P4, Canada
Received 2 February 2009; revised 5 April 2009; accepted for publication 6 April 2009;
published 8 May 2009
Brachytherapy is a mature treatment modality that has benefited from technological advances.
Treatment planning has advanced from simple lookup tables to complex, computer-based dosecalculation algorithms. The current approach is based on the AAPM TG-43 formalism with recent
advances in acquiring single-source dose distributions. However, this formalism has clinically
relevant limitations for calculating patient dose. Dose-calculation algorithms are being developed
based on Monte Carlo methods, collapsed cone, and solving the linear Boltzmann transport equation. In addition to improved dose-calculation tools, planning systems and brachytherapy treatment
planning will account for material heterogeneities, scatter conditions, radiobiology, and image
guidance. The AAPM, ESTRO, and other professional societies are working to coordinate clinical
integration of these advancements. This Vision 20/20 article provides insight into these
endeavors. 2009 American Association of Physicists in Medicine. DOI: 10.1118/1.3125136
Key words: brachytherapy, treatment planning
I. HISTORY OF BRACHYTHERAPY DOSIMETRY
Many of our technological discoveries are distinctly marked
in history. Independent of each other, two of these discoveries launched the start of the radiation oncology era. Wilhelm
Rentgen discovered x rays in November 1895, and shortly
afterward, Henri Becquerel accidentally exposed a photographic plate to uranium in 1896, identifying the phenomenon of emitted radiation.1 The work of Pierre and Marie
Curie in 1898 was needed to extract radium from pitchblende
ore to determine the origin of this penetrating radiation. Very
soon after these first steps, new pathways were explored to
apply these radiations in the first treatments of patients. Becquerel himself experienced the effects of radiation exposure
by carrying a tube containing decigrams of radium chloride
in his vest pocket. He recorded the progression of his skin
reaction.2 The first medical experiences belong to Danlos and
Bloch3 in 1901 of Paris and Abb4 in 1904 of New York. The
Radium Biological Laboratory was created in Paris in 1906,
and Finze in London started treatments with radium in 1909.
A book on radium therapy, which is now known as brachytherapy, was published in 1909 by Wickham and Degrais.5
These early 20th century achievements established the medical application of radiation within one decade of its discovery. The first investigators were by necessity led by clinical
observations and developed from their experiences the rules
to avoid errors in clinical applications. In medical science,
such processes soon lead to systems or schools. For brachytherapy, 226Ra was the only radionuclide for these systems.6
The basic principles of its use in brachytherapy were estab2136
0094-2405/2009/366/2136/18/$25.00
2136
2137
2137
nal x rays, and ii summation of dose patterns of the individually reconstructed sources. The conventional systems
were applicator based, not anatomy based. They did not utilize the anatomical information, so the relation between dose
deposition and the tumor volume as well as the volume of
healthy tissue and organs at risk OARs was underdeveloped. Implementation of modern resources with accurate calculation techniques and imaging procedures allows us to increase the efficacy of brachytherapy in terms of clinical
outcome by demonstrating whether or not a chosen isodose
covers the clinical target volume CTV and spares OARs.
Patient-specific treatment planning is presently considered
useful in our treatments and is essential for achieving the
treatment planning standards of modern radiation oncology
in more complicated cases.
I.B. Prescription dose
2138
2138
FIG. 2. Coordinate system for the AAPM TG-43 brachytherapy source dosimetry formalism from Rivard et al. Ref. 24.
FIG. 1. The escargot diagram in the Paris system is a tool for manual
calculation of the dose rate at points at distances in the plane transversal to
the source. The curves represent the dose rate in cGy h1 for sources of 1
up to 7 cm length and a linear strength of 1 mR h1 m2 cm1
8.7 Gy h1 m2 cm1. From Dutreix et al. Ref. 20.
tion such as bone and lung. Then, these values must be taken
from textbooks or other resources where it is easy to find
great 25% disparities in the tabulated values.21 This is
apparent when trying, for example, to list values from the
literature for a radionuclide with a complex photon spectrum
such as 192Ir. These flaws may lead to errors, for example,
when the effective activity is measured from dose rate in air
by the vendor while the user of a given calculation system
has included a different value. The fact that this concern
is not imaginary but very real is demonstrated in the IAEA
survey on misadministrations in medical radiation applications, often occurring in coordination with a lack of
training and supervision of the personnel involved in the
procedures.22
As the quantities for A, , and Fm in Eq. 1 are not in
accordance with the SI system, and therefore considered obsolete, Eq. 1 needs to be rephrased for the dose rate to
r. A very detailed step-by-step description is
water D
W
given in the recent book by Baltas et al.23 resulting in Eqs.
2 and 3,
1 g en
r = K
D
W
R
r = S 1 g en
D
W
K
r0
r
1
r
f as,Wr,
f as,Wr.
2139
2139
r = S r0
D
K
r
g Pr anr,
r, = S GLr, g r Fr, .
D
K
L
GLr0, 0
2140
2140
description is provided of the main publicly accessible archives or databases of dosimetry datasets to provide guidance on dosimetry parameter choice. Toward providing guidance on dataset choice, recommendations to medical
physicists by Rivard et al.37 are presented below:
a
b
c
Due to the simplicity of brachytherapy dosimetry in comparison to external beam dosimetry, a basic understanding of
radiological physics interactions can generally explain five
limitations of the AAPM TG-43 dosimetry formalism.
For multisource clinical applications, ISA may be significant. Applicator:radiation interactions and applicator shielding may detract from the accuracy of conventional TG-43
based dose calculations.44 For radiation shielding between
2141
sources or by an applicator, the attenuation of water is replaced with a high-Z material. For LDR low-energy photonemitting sources, this high-Z material may for example be a
silver Z = 47 radio-opaque marker used for identification
during postimplant CT localization. For HDR high-energy
photon-emitting sources, the material may be a stainless steel
applicator Z = 26. Using Ag = 10.5 g / cm3 and Fe = 7.8 g /
cm3 with a minimal thicknesses of 0.1 mm and negligible
water attenuation, the high-Z photon attenuations are given
in Fig. 4. As in the prior two examples, the photoelectric
effect is largely responsible for the differences in comparison
to water.
III.A.4. Differences between radiation scattering for
dataset acquisition and patient treatment
The effect of phantom dimensions and volume in MC
dosimetry has been reported in literature as early as
1991.43,4549 For high-energy photon-emitting brachytherapy
sources such as 192Ir, dose differences greater than 5% are
possible within 10 cm of the source, as shown in Fig. 5.
However, studying varying phantom dimensions quantifies
only the effect of excess material or missing backscatter
close to the phantom boundary as source position is fixed
and phantom radius decreases. Therefore, as argued and
shown in Pantelis et al.50 and later by Lymperopoulou et
al.,51 corresponding findings cannot be readily translated to
the effect expected in actual clinical practice where a source
dwell position is programmed close to the patient contour as
scattering conditions are altered in total, including lateral
scatter.
Further, the proportion of dose due to primary and scattered radiation significantly changes as a function of distance
from the source, as shown in Fig. 6, which is also for 192Ir.52
2141
FIG. 6. Components of 192Ir dose as a function of radial distance. Reproduced with permission from R. E. Taylor and D. W. Rogers, Med. Phys. 35,
4933 2008. Copyright 2008, American Association of Physicists in
Medicine.
2142
2142
TABLE I. Sensitivity of commonly treated anatomic sites to dosimetric limitations of the current brachytherapy
dose calculation formalism. Items flagged as Y indicate the authors opinion that significant differences
between administered and delivered dose are possible due to the highlighted dosimetric limitation.
Anatomic site
Prostate
Breast
GYN
Skin
Lung
Penis
Eye
Source energy
Absorbed dose
Attenuation
Shielding
Scattering
Beta/kerma dose
High
Low
High
Low
High
Low
High
Low
High
Low
High
Low
High
Low
N
Y
N
Y
N
Y
N
Y
N
Y
N
Y
N
Y
N
Y
N
Y
N
Y
N
N
N
Y
N
N
N
Y
N
Y
N
Y
Y
N
Y
Y
N
N
N
N
Y
Y
N
N
Y
N
N
N
Y
Y
Y
Y
Y
Y
Y
Y
N
N
N
N
N
N
N
N
Y
N
N
N
Y
N
2143
tissues, and conventional TPS cannot account for this shielding. Again, absorbed dose is underestimated by several percent for low-energy brachytherapy sources. Lung implants
typically include tissues with mass densities three to four
times less than conventional tissues. Consequently, scattering
conditions for both high- and low-energy brachytherapy
sources are not congruent between the clinical implant and
the planned approach. Absorbed dose and attenuation are
also significantly different for low-energy sources, and dose
calculations close to high-energy sources such as HDR 192Ir
can be in error by several percent within a few millimeters of
the source. Brachytherapy of the penis is performed using
either HDR high-energy sources or LDR low-energy sources.
Both applications overestimate administered dose due to radiation scattering conditions. Further, low-energy sources underestimate dose due to differences in tissue composition between water and tissue. Eye plaque brachytherapy has been
conducted for several decades but is sensitive to all of the
aforementioned dosimetric limitations.
In summary, it is clear that most applications of brachytherapy are subject to limitations of the current dosecalculation formalism. Thus, it is imperative that advances be
made to resolve these discrepancies and provide clinical users a realistic depiction of individualized brachytherapy dose
administration.
IV. BRACHYTHERAPY TREATMENT PLANNING
RESEARCH
IV.A. Introduction
2143
2144
rate calculations of the multiple-scatter components necessitate the full 3D geometry, especially in the presence of material inhomogeneities for which the interaction cross
sections can vary greatly.
An early implementation of a convolution method for
brachytherapy has been demonstrated by Williamson et al.92
In this approach, the multiple-scatter component is provided
by precomputed MC dose spread array. For 125I and 137Cs
point-source dose calculations, the results were shown to be
within 3% of the MC calculations with a gain in speed of
factors of 2050 at that time. In this approach, heterogeneity
corrections were accounted for by rescaling the dose spread
of the once-scattered photons. Another approach proposed by
Williamson et al.93 used a scatter-subtraction method based
on 1D scatter integration. While being about 1000 times
faster than MC, it was limited to water-equivalent heterogeneities for cylindrical geometries. This method was further
extended to a 2D scatter integration,94 allowing arbitrary geometry. However, its heterogeneity correction was limited by
analytical correction factors for a wide range of atomic numbers and material densities.95 The latter incarnation was
shown to be 50 000 100 000 times faster than MC while
maintaining an accuracy of 10% except for low-energy photons and high-atomic number heterogeneity materials. However, in this approach multiple scattering is neglected and the
heterogeneity geometry was limited to a thin slab.
The application of a CC algorithm in brachytherapy has
been pursued for many years by the Helax/Nucletron group
in Uppsala, Sweden. With this algorithm, a successivescattering superposition method has been developed in which
the first-scatter dose distribution is used to derive the higher
order multiple-scatter distributions. In this formalism, the
multiple-scatter kernel can be isotropic for low-energy
photons96 or when oriented along the primary photon.80 A
scaling method was also developed for the successivescattering superposition in the presence of high-Z and highdensity materials.97 A source description formalism was also
proposed in the context of primary and scatter dose separation PSS formalism that is backward compatible with the
current TG-43 dose-calculation formalism and is used as input into the CC algorithm.98 CC performance was recently
studied for relevant brachytherapy geometries and for 28, 60,
and 350 keV sources. A total of 31 800 voxels/ s 0.2 mm3
voxel, 52 directions were calculated for permanent prostate
seed geometry.90 The authors further tested implementation
on parallel hardware, specifically GPU cards, and showed it
to be 100 times faster for the same geometry.90
Finally, Wang and Sloboda99,100 also proposed a formalism of primary, once-scatter and multiple-scatter separation.
The novelty is that the once-scatter is evaluated using a microbeam ray-tracing algorithm, which includes basic interaction physics such as Compton, Rayleigh, and photoelectric
effect. The advantage of this approach is that the once-scatter
contribution does not have to be scaled to account for material heterogeneities.99 The multiple-scatter dose is obtained
from a nonlinear curve fitting of MC multiple-scatter dose in
various heterogeneous media.100 Accuracy was within 10%
for all cases studied.
Medical Physics, Vol. 36, No. 6, June 2009
2144
Monte Carlo uses stochastic approaches random numbers to sample the probability density functions describing
the phenomena underlying the transport of particles through
matter for simulations. With sufficient statistics or particle
histories, MC obtains precise solutions to a variety of problems in radiation therapy. Another calculation approach consists of directly solving the linear Boltzmann transport equation LBTE through deterministic means. Two general
approaches with application to brachytherapy can be seen in
the literature: solving the differential LBTE81,101 or the integral formulation.102 The LBTE is commonly solved by discretization of the parameters phase space. The most commonly seen method in scholarly articles is the angular
domain or the use of DO. Discretization is also performed in
space finite difference or finite element and in energy with
appropriate multigroup cross sections. The system can be
coupled to handle neutral, charged, and coupled photonelectron-positron transport for iterative solutions. The discretization of the space dimension is well suited to problems
in radiation therapy where voxel-based geometries of patients are constructed from tomographic images.101
It is generally understood that the accuracy of deterministic approaches is directly related to discretization,101,103
with fine steps leading to accurate solutions at the price of a
larger system of equations to be solved. Thus, the technique
is numerically intensive. As noted by Daskalov et al.,101 the
deterministic concept of accuracy is not the same as for MC
dose calculation and is related to a systematic difference between the solution and the truth. With deterministic tools,
uncertainty does not have a stochastic component, while MC
uncertainties have stochastic and systematic components. An
important issue for brachytherapy is the ray effect, which
results from the nonphysical fluence buildup due to the limited number of angles for near pointlike sources in lowdensity media. To diminish this artifact, stochastic or semianalytic methods are generally used to determine the oncescattered dose distribution within the discretized phase
space.81,101
The first application of a deterministic approach to HDR
brachytherapy used a finite-difference multigroup-DO solver
from Los Alamos National Laboratory LANL called
DANTSYS.104 Though limited to 2D, the results were within
2 of EGS4 MC calculations.101 This approach was further
extended, namely, by reducing the energy group cross sections, to calculation of the absorbed dose rate around a 125I
brachytherapy seed for a 2D cylindrical geometry. The deterministic method was shown to be 100-fold more efficient
than the EGS4 MC code.105
Zhou and Inanc102 introduced another approach to solve
the integral representation of the LBTE and studied ISA in
125
I seed implants. The algorithms were extended to account
for fluorescence x rays, and parallelization was introduced.
Single seeds and more complex configurations such as the
Quality Assurance Review Center www.qarc.org geometry
for clinical protocol accreditation were studied.106 The calcu-
2145
2145
2146
potential solution.114116 Still, some guidelines must be provided to the end user, otherwise large variations would result
when using MC methods.
V. PREDICTIONS AND RATIONALE FOR
BRACHYTHERAPY TPS TRENDS
The previous sections describe the past, present, and future of brachytherapy dosimetry in a narrow interpretation of
the term dosimetry as a dose-calculation procedure. We followed the history from the systems of the precomputer era,
from the table or nomogram based manual systems, via the
classical Sievert-based dose calculation Sec. I D, to the
present-day TG-43 based system Sec. II, toward the complex algorithms for primary-scatter separation superposition/
convolution models and the MC approach Sec. IV D. We
now summarize a number of activities by committees within
the AAPM and jointly with other professional societies to
help shape the future of brachytherapy dosimetry; here used
in the wider interpretation of the term. A detailed uncertainty
analysis will help identify areas where the largest benefits
can be achieved from research activities. Easily accessible
traceable source strength calibration methods, preferably in
the quantity dose to water are helpful and reassuring for inhouse check procedures of source deliveries. We must strive
for a situation where dose is interpreted unequivocally
among institutions, for the same starting points whichever
system is used. Easily accessible consensus datasets for
source characterization, both in TG-43 format and in the
more complex algorithms, are then required for commissioning TPS and for quality control. Maybe the most important
evolution in brachytherapy dosimetry is the consequence of
modern imaging technologies. Section V presents our views
on a number of these issues.
V.A. Working groups and joint international
coordination
2146
2147
2147
To be useful in the clinic, MC TPS may come with predefined CAD based applicator geometries and physical properties. We expect that source modeling and its associated
phase-space file will be mandatory as per a new AAPM prerequisite. These might be specific to the type of MC code
implemented by the commercial vendor, but would still have
to meet stringent validation, akin to the AAPM brachytherapy dosimetry prerequisites. As we expect most of the
applicators, catheters, and needles to become MRI/CT compatible, artifacts will become less of an issue. However, image processing will be implemented to minimize streaking of
permanent seed implants.
It is expected that the next generation algorithms will be
able to deal with a number of the problems related to the
accuracy of dose calculation discussed in Sec. III. The influence of tissue density and tissue composition effects on the
resulting dose distribution was discussed in detail in the recent AAPM Anniversary Paper on Brachytherapy Physics by
Thomadsen et al.16 While the reader is referred to that document, we summarize that a direct MC calculation approach
can address the substantial dose effects of several percent
possible with tissue composition changes such as soft tissue
calcifications,61 adipose, and fatlike tissues123 depending on
the photon energy. The challenge remains in the design of
imaging technology to allow advanced brachytherapy TPS to
account for the relevant spatial and density information. Applicator and catheter shielding effects have similar influence
on the dose deposition.124,125 Further, those involved with
TPS algorithm development would benefit if vendors provide
a full description of their applicators. This is one step beyond
the tools currently available for full applicator geometry description for reconstruction, viewing, and manipulation in
3D. An open database to share the information on material
composition in addition to a series of systematic MC research studies on the influence of a wide spectrum of photon
radiation on dose is welcomed. Brachytherapy TPS development should be less vendor specific and instead provide compatibility with afterloading equipment from other vendors.
Standardization efforts for brachytherapy DICOM-BT by
the cross-societal Integrating the Healthcare Environment for
Radiation Oncology IHE-RO taskforce are underway.
There are more problems that can be addressed in the
coming years. One is a systematic and comprehensive study
on the influence of secondary photon radiation on dose from
beta components e.g., bremsstrahlung of radionuclides used
or proposed for brachytherapy. Often the beta particles themselves or low-energy photons are absorbed in the source ma-
2148
2148
2149
2149
2150
2150
radium therapy, Am. J. Roentgenol., Radium Ther. Nucl. Med. 70, 739
740 1953.
18
W. J. Meredith, Radiotherapeutic physics: V. Dosage control in interstitial gamma-ray therapy, Br. J. Radiol. 24, 380384 1951.
19
R. J. Shalek and M. A. Stovall, in Radiation Dosimetry, edited by F. H.
Attix and E. Tochlin Academic, New York, 1969, Vol. 3, pp. 776798.
20
A. Dutreix, G. Marinello, and A. Wambersie, Dosimtrie en Curiethrapie Masson, Paris, 1982.
21
S. Jayaraman, L. H. Lanzl, and S. K. Agarwal, An overview of errors in
line source dosimetry for gamma-ray brachytherapy, Med. Phys. 10,
871875 1983.
22
IAEA Report No. 17 IAEA, Vienna, 2000.
23
D. Baltas, L. Sakelliou, and N. Zamboglou, The Physics of Modern
Brachytherapy for Oncology Taylor & Francis, New York, 2007.
24
M. J. Rivard, B. M. Coursey, L. A. DeWerd, W. F. Hanson, M. S. Huq, G.
S. Ibbott, M. G. Mitch, R. Nath, and J. F. Williamson, Update of AAPM
Task Group No. 43 Report: A revised AAPM protocol for brachytherapy
dose calculations, Med. Phys. 31, 633674 2004.
25
J. F. Williamson, Recent developments in brachytherapy physics, Proceedings of the ABS 1994 Summer School Medical Physics, Madison,
WI, 1995.
26
J. F. Williamson, Monte Carlo and analytic calculation of absorbed dose
near 137Cs intracavitary sources, Int. J. Radiat. Oncol. Biol. Phys. 15,
227237 1988.
27
J. F. Williamson, The Sievert integral revisited: evaluation and extension
to 125I, 169Yb, and 192Ir brachytherapy sources, Int. J. Radiat. Oncol.,
Biol., Phys. 36, 12391250 1996.
28
J. F. Williamson, Monte Carlo-based dose-rate tables for the Amersham
CDCS.J and 3M model 6500 137Cs tubes, Int. J. Radiat. Oncol. Biol.
Phys. 41, 959970 1998.
29
R. van der Laarse, presented at Proceedings of the Seventh Intern Conference on the Use of Computers in Radiation Therapy, Japan, 1980 unpublished.
30
R. Nath, L. L. Anderson, G. Luxton, K. A. Weaver, J. F. Williamson, and
A. S. Meigooni, Dosimetry of interstitial brachytherapy sources: Recommendations of the AAPM Radiation Therapy Committee Task Group No.
43. American Association of Physicists in Medicine, Med. Phys. 22,
209234 1995.
31
R. Paterson, A dosage system for gamma ray therapy. Part I. Clinical
aspects, Br. J. Radiol. 7, 592612 1943.
32
H. Parker, A dosage system for gamma ray therapy. Part II. Physical
aspects, Br. J. Radiol. 7, 612632 1943.
33
B. Pierquin and G. Marinello, A Practical Manual of Brachytherapy
Medical Physics, Madison, WI, 1997.
34
W. M. Butler, W. S. Bice, Jr., L. A. DeWerd, J. M. Hevezi, M. S. Huq, G.
S. Ibbott, J. R. Palta, M. J. Rivard, J. P. Seuntjens, and B. R. Thomadsen,
Third-party brachytherapy source calibrations and physicist responsibilities: Report of the AAPM Low Energy Brachytherapy Source Calibration
Working Group, Med. Phys. 35, 38603865 2008.
35
S. M. Seltzer, P. J. Lamperti, R. Loevinger, M. G. Mitch, J. T. Weaver,
and B. M. Coursey, New national air-kerma-strength standards for 125I
and 103Pd brachytherapy seeds, J. Res. Natl. Inst. Stand. Technol. 108,
337358 2003.
36
M. J. Rivard, W. M. Butler, L. A. DeWerd, M. S. Huq, G. S. Ibbott, A. S.
Meigooni, C. S. Melhus, M. G. Mitch, R. Nath, and J. F. Williamson,
Supplement to the 2004 update of the AAPM Task Group No. 43 Report, Med. Phys. 34, 21872205 2007.
37
M. J. Rivard, C. S. Melhus, and J. F. Williamson, Brachytherapy dose
calculation formalism, dataset evaluation, and treatment planning system
implementation, in Clinical Dosimetry for Radiotherapy: AAPM Summer School, edited by D. W. O. Rogers and J. E. Cygler Medical Physics,
Madison, WI, 2009.
38
L. A. DeWerd, M. S. Huq, I. J. Das, G. S. Ibbott, W. F. Hanson, T. W.
Slowey, J. F. Williamson, and B. M. Coursey, Procedures for establishing and maintaining consistent air-kerma strength standards for lowenergy, photon-emitting brachytherapy sources: Recommendations of the
Calibration Laboratory Accreditation Subcommittee of the American Association of Physicists in Medicine, Med. Phys. 31, 675681 2004.
39
Z. Li, R. K. Das, L. A. DeWerd, G. S. Ibbott, R. W. Kline, A. S. Meigooni, J. Perez-Calatayud, M. J. Rivard, R. S. Sloboda, and J. F. Williamson, Dosimetric prerequisites for routine clinical use of photon emitting
brachytherapy sources with average energy higher than 50 keV, Med.
Phys. 34, 3740 2007.
2151
40
63
2151
2152
77
2152
2153
HDR brachytherapy of the breast, accounting for the effect of finite patient dimensions and tissue inhomogeneities, Med. Phys. 33, 45834589
2006.
126
J. J. DeMarco, R. E. Wallace, and K. Boedeker, An analysis of MCNP
cross-sections and tally methods for low-energy photon emitters, Phys.
Med. Biol. 47, 13211332 2002.
127
A. Gerbaulet, R. Ptter, J.-J. Mazeron, H. Meertens, and E. Van Limbergen, The GEC ESTRO Handbook of Brachytherapy ESTRO, Brussels,
2002.
128
R. Nath, L. Anderson, D. Jones, C. Ling, R. Loevinger, J. Williamson,
and W. Hanson, AAPM Report No. 21, 1987.
129
NCS Report No. 4, 1991.
130
H.-J. Selbach, H.-M. Kramer, and W. S. Culberson, Realization of reference air-kerma rate for low-energy photon sources, Metrologia 45,
422428 2008.
131
A. Herskovic, S. Lee, and T. Padikal, Utilization of the computed tomography scanner in interstitial dosimetry, Radiology 135, 781782
1980.
132
B. Pierquin and J. V. Fayos, Dosimetry by tomography in interstitial
curietherapy: Point technique, Am. J. Roentgenol., Radium Ther. Nucl.
Med. 87, 585592 1962.
133
C. C. Ling, M. C. Schell, K. R. Working, K. Jentzsch, L. Harisiadis, S.
Carabell, and C. C. Rogers, CT-assisted assessment of bladder and rectum dose in gynecological implants, Int. J. Radiat. Oncol., Biol., Phys.
13, 15771582 1987.
134
S. L. Schoeppel, M. L. LaVigne, M. K. Martel, D. L. McShan, B. A.
Fraass, and J. A. Roberts, Three-dimensional treatment planning of intracavitary gynecologic implants: Analysis of ten cases and implications
for dose specification, Int. J. Radiat. Oncol., Biol., Phys. 28, 277283
1994.
135
L. Feuvret, G. Noel, J. J. Mazeron, and P. Bey, Conformity index: A
review, Int. J. Radiat. Oncol., Biol., Phys. 64, 333342 2006.
136
H. H. Holm, N. Juul, J. F. Pedersen, H. Hansen, and I. Stroyer, Transperineal 125iodine seed implantation in prostatic cancer guided by transrectal ultrasonography, J. Urol. Paris 130, 283286 1983.
137
J. C. Blasko, P. D. Grimm, and H. Ragde, Brachytherapy and organ
preservation in the management of carcinoma of the prostate, Semin.
Radiat. Oncol. 3, 240249 1993.
138
A. A. Martinez, I. Pataki, G. Edmundson, E. Sebastian, D. Brabbins, and
G. Gustafson, Phase II prospective study of the use of conformal highdose-rate brachytherapy as monotherapy for the treatment of favorable
2153