★阿修羅♪ > 原発・フッ素19 > 910.html
 ★阿修羅♪  
▲コメTop ▼コメBtm 次へ 前へ
NATO NATO/PFP UNCLASSIFIED Pre-released RTO Technical Report
http://www.asyura2.com/11/genpatu19/msg/910.html
投稿者 SaveChild厨 日時 2012 年 7 月 11 日 03:35:28: hfhZnjAARar8w
 

(回答先: Re: 宇宙飛行士が内服している薬・食品について 爺さんより 投稿者 SaveChild厨 日時 2012 年 6 月 07 日 18:48:26)

NATO/PFP UNCLASSIFIED
Pre-released RTO Technical Report
Radiation Bioeffects and Countermeasures
http://www.usuhs.mil/afrri/outreach/pdf/$$TR-HFM-099-Pre-Release-ALL.pdf
----------------------------------------------

コピー・整形がうまくできていない部分があります。上記原文PDFで引き合わせしてください。

This report has been pre-released, in its original format, to make it immediately vailable to the scientific community. Once the report has been edited, formatted and

formally approved, the RTA will publish an official version under reference .

NORTH ATLANTIC TREATY ORGANISATION RESEARCH AND TECHNOLOGY ORGANISATION

AC/323() www.rta.nato.int

Executive Summary
William F. Blakely (USA)
RTG-033 Chair

There are hundreds of instances in which one or more persons were accidentally overexposed to ionizing radiation. A radiological or nuclear attack is also a possibility.

Because of recent terrorist activities and intelligence information, there is strong sentiment that it is not a question of if, but when, a radiological or nuclear terrorist

attack will occur. These radiation hazards have the potential of disrupting and compromising NATO military operations, and therefore represent a current and creditable

threat issue. The objectives of the RTG-033 are to develop the scientific basis for new and improved methods to prevent, assess, treat, and manage casualties and long-

term health effects associated with ionizing radiation exposure from evolving threats in military operations.

The RTG-033 consisted of participation from NATO member (10) and partner (1) nations. It was organized into 4 subgroups: 1) radiobiology mechanisms and late

effects, 2) radiation injury assessment and biodos-imetry, 3) prophylaxis and therapy, and 4) combined injury and treatment. Each subgroup selected co-chairs, who

developed specific aims and tasks for the 3-year activity. The RTG-033 members met at four annual meetings, hosted by the RTG-033 member research Institutes and

nations. The 2005 kickoff meeting at the Armed Forces Radiobiology Research Institute (USA) produced a meeting report CDROM that included 22
manuscripts and presentations (see Appendix).
Subgroup #1 focused on basic radiobiology relevant research topic reports that addressed: a) new research models to investigate late effects of radiation (Chapter 2), b)

low-level radiation bioeffects and their mechanisms (Chapter 3), c) use of gene expression technology to support investigations in radiobiological mechanisms (Chapter

4), and d) review of use of molecular biomarkers for assessment of the acute radiation syndrome (ARS) sub-syndromes (Chapter 5).

Subgroup #2 final report consists of several components. First, the Co-chairs provide a summary report of the overall Subgroup products including the development of

improved methods for casualty assessment (Chapter 6). A major highlight of this group’s activities includes an evaluation tool and process to facilitate assessment
of the selection, advancement, and of emerging biodosimetry methodologies. Additional products included reports on: i) biodosimetry use during a radiological mass-

casualty incident (Chapter 7) and ii) progress on automation of the ―gold standard? cytogenetic chromosome aberration-based assay (Chapter 8).

Subgroup #3 contributions to the final report address research progress and use of countermeasures for protection, treatment, and management of radiological

casualties (Chapters 9?11).

Subgroup #4 provides an updated review of combined injuries and treatment (Chapter 12).

Appendix materials include annual meeting agendas, subgroup bibliographies, technical reference level (TRL) guidance, and the 2005 annual meeting proceedings in

CDROM format.



Table of Contents
Page
Executive Summary
Table of Contents
Abstract and Keywords
Foreword
Terms of Reference
Acknowledgements
RTG-033 Members
RTG-033 Final Report Contributors
1 Chapter 1 ? Summary of RTG-033 activities n
2 Chapter 2 ? Development of models to study radiation-induced late effects n
3 Chapter 3 ? Stimulation of the natural anti-tumor cells by single or fractionated
irradiation of mice with x-rays n
4 Chapter 4 ? German contributions using gene expression studies to detect
radiation targets and to discriminate radiation associated tumors from other
ethiologies n
5. Chapter 5 ? Molecular biomarkers of radiation injury and acute radiation syndrome n
6. Chapter 6 ? Radiation injury assessment and biodosimetry n
7 Chapter 7 ? Early biodosimetry response: recommendations for mass-casualty radiation accidents and terrorism n
8 Chapter 8 ? High-throughput robotic blood handling and cytogenetic sample preparation n
9 Chapter 9 ? Progress in radiation countermeasures n
10 Chapter 10 ? Advances in cytokine-based treatment n
11 Chapter 11 ? Radiation injury treatment and prophylaxis: relevant options for an Radiation Dispersal Device scenario n
12 Chapter 12 ? Radiation in combination with trauma, infectious disease, or chemical exposures n
13 Appendix materials n
13.1 Annual meeting agendas n
13.1.1 June 2005 RTG-033 annual meeting agenda n
13.1.2 June 2006 RTG-033 annual meeting agenda n
13.1.3 June 2007 RTG-033 annual meeting agenda n
13.1.4 June 2008 RTG-033 annual meeting agenda n
13.2 Subgroup bibliography n
13.2.1 Subgroup#2 bibliography n
13.2.2 Subgroup#3 bibliography n
13.3 Technical reference level guidance n
13.3.1 NATO technology readiness levels guidance n
13.3.2. U.S. Department of Defense TRL guidelines for pharmaceuticals
and medical devices n
13.3.3 U.S. Department of Defense PCR assay and ELISA guidelines n
13.3.4 U.S. Department of Health and Human Services guidelines for
medical TRLs n
13.4 Meeting proceedings n
13.4.1 June 2005 RTG-033 meeting proceedings ? CDROM n
13.4.2 June 2005 RTG-033 meeting proceedings ? PDF (manuscripts only) n

Abstract and Keywords

Abstract

The Radiation Bioeffects and Countermeasures RTG-033 Final Report addresses: i) radiobiology mechanisms related to acute and late radiation effects to high and low doses, ii) improved methods to assess radiation
injuries, iii) an update on the use of medical countermeasures to present, treat, and manage radiation casualties and long-term health effects, and iv) combined injury and treatment. Projected deliverables, established at the
onset and taking into account military relevant research and operational needs, were accomplished.

Keywords

Radiation bioeffects, mechanisms, late effects, biodosimetry, radiation injury assessment, medical recording tools, acute radiation syndrome, medical countermeasures, cytokine therapy, combined injury.

Foreword

The Human Factors and Medicine Panel of the NATO’s Research and Technology Organization (RTO/HFM) has long recognized the risks of radiological threats to military forces. Indeed, significant gaps still exist in our
understanding of basic mechanisms of radiation-induced biological effects, threat assessment capabilities applicable, and medical countermeasures to protect, mitigate, treat, and manage acute- as well as late radiation
effects for military operations. The RTO/HFM panel has supported the sustainment of radiobiology-based RTG’s for over 2 decades.

The Radiation Bioeffects and Countermeasures Task Group HFM-099/RTG-033 was organized and initiated in June 2005 after a gap of two years relative to the activities of an earlier radiobiology HFM-046/RTG-006.
The USA was asked to staff and provided the leadership (Chair) for the RTG-033 that was comprised of NATO member (~10) and partner (1) nation representatives. The RTG activities were accomplished by the
cooperative efforts of the RTG-033 members during the 3-yr activity; the co-chairs for the subgroups were drawn from seven nations and contributors to this final report were drawn from both RTG members and non-
member expert collaborators. I commend the RTG-033 for their efforts and the valuable content of their Final Report.

As reflected by the content of the RTG-033 Final Report, there has been considerable progress in radiobiology research to merit this activity. No single nation is equipped to undertake these immense tasks to fill critical
radiobiology gaps applicable for operations of NATO forces. The RTG-033 has provided a valuable venue to convene radiobiology expert scientists from a broad spectrum to share relevant knowledge. Current events and
risk assessment of the evolving threats underscore the importance to continue these activities.

Dr. Marek K. Janiak (POL)
RTG-033 Referee

Terms of Reference
RESEARCH TASK GROUP 033
RADIATION BIOEFFECTS AND COUNTERMEASURES HUMAN FACTORS AND MEDICINE (HFM) PANEL

I. ORIGIN
A. Background
1. Proliferation of radioactive material, nuclear weapons and nuclear power facilities has increased the likelihood that multinational military forces will encounter a local or

widespread radio-logical hazard. Such nuclear/radiological hazards can result from hostile acts short of nuclear war to the deliberate spread of radioactive material from

industrial or medical sources, accidental or intentional destruction of nuclear facilities during a conflict, accidents involving radiation sources, or the detonation of improvised nuclear devices by terrorists or hostile forces.
2. These radiation hazards have the potential of disrupting and compromising NATO military operations, and therefore represent a current and creditable threat issue.
3. Recommended countermeasures must take into account operational as well as medical implica-tions of exposure to ionizing radiation over a sizable range of exposure

intensities, qualities, and durations. Consideration of this potential variation in radiation exposure intensities is in part captured by the NATO STANAG 2083 and its listing

of radiation exposure status (RES)

categories ranging 1 through 3.
4. Of additional concern, is that the nuclear/radiological hazard is amplified by the proliferation of the weapons of mass destruction, biological and chemical threat

agents, and other environmental stressors, that may present during a mission.
5. Previous research study groups (NATO RSG-23) on ionizing radiation injury have concentrated almost exclusively on the dosimetry and biological effects of the high

doses associated with general nuclear war. The management of early acute effects of such radiation exposure, continued operational availability of exposed personnel and

ultimately survival predominated.
The changing world order and consequent defense planning assumptions do not exclude high dose exposure (though in different circumstances) but predicate a new

emphasis on the lower doses that may be encountered in future operations whether through accident or hostile intent.
These exposures may produce some acute effects that may impact on operational performance, but the principal risk will be in long-term effects. 6. Thorough evaluation

of major late-arising pathologies (e.g., cancer, fibrosis, chronic recurrent infections) in terms of pathogenesis and the identification, development and testing of safe and
effective medical countermeasures requires considerably more time than the three year study cycle allotted by the NATO RTO.
7. Despite the successes of previous research task groups in developing essential procedures and equipment requirements to manage the extreme radiological hazards

associated with nuclear weapons in the context of general war, the reduced risk of general nuclear war has made past ssumptions about acceptable levels of radiological

contamination and acceptable doses of radiation to soldiers inadequate. Although these exposure levels and associated health hazards have been revisited (by virtue of

the NATO ACE Directive 80-63), justified, and considered relative to currently acceptable peacetime limits for occupational/non-occupational workers (by virtue of the

NATO ACE Directive 80-63), still additional reevaluation and refinement of these radiation exposure guidelines are needed, particularly in light of the fact that current

exposure guidelines do not take into account the effects of exposure rate, radiation quality and committed dose.
8. The need to research and develop new and improved methods to counter these nuclear/ radio-logical threats is clear and unambiguous. The current iotechnical/bioengineering revolution is presenting unprecedented opportunities to significantly extend our understanding of the molecu-lar and cellular details of

radiation toxicity, thus enhancing our capacity for strategic design and development of safe and effective pharmacologic/biologic countermeasures.
9. The Research Task Group, TG006 made significant progress toward developing essential scien-tific bases upon which to build new bioassessment tools and to develop

new and better methods for preventing and treating injuries associated with ionizing radiation exposure. This work war-rants a continuation of effort, thus a new three

year study cycle is proposed.
B. Justification
10. Medical and armament planners require scientifically founded data to develop guidance and recommendations in order to deal with the new situation described above.

Recommendations must take into account operational as well as medical implications of exposure to ionizing radiation at a range from RES category 1 through 3 STANAG

2083). Similarly, planners need to consider the radiological guidelines set forward in the ACE Directive 80-63. Further, due to the proliferation of the weapons of mass

destruction, interactions with BW/CW agents, medical prophylaxis, as well as other stressors must also be addressed.
11. It is therefore clear that currently available physical and biomedical scientific data are not sufficient to develop the necessary advice for commanders in emergency

situations, nor are doctrine, training, and equipment. The findings and recommendations will be passed on to the appropriate groups responsible for preparing guidelines

for operations in low-level radiation environments.
12. The proposed new Task Group is required because no member nation has the expertise and resources in all these areas of interest; therefore, a cooperative research

and study effort is necessary and should address the topics as listed below.
II. OBJECTIVES
A. Research Area and Scope
13. The research area and scope of the proposed RTG is defined by the group’s mission, namely ―To develop the scientific basis for new and improved methods to

prevent, assess, treat, and manage casualties and long-term health effects associated with ionizing radiation exposure from evolving threats in military operations?.
B. Specific Goals
14. Goals include: (a) development of broad-spectrum bioassessment tool(s) for sensitive, accurate, and reliable detection of radiation-associated injuries; (b)

stablishment of underlying bases of health-compromising, performance-decrementing radiation injuries at the molecular, cellular and organ-system levels; and (c) based

on established origins and mechanisms of radiation-induced pathology-induction, development of effective protocols for the prevention and treat-ment of those initial

radiation injuries that would otherwise cascade into overt disease, com-promising health and performance of personnel so affected.
15. The research topics to be covered by the RTG will encompass the following: (a) prototype development and testing of candidate biodosimetry assessment tools for

radiation exposures of varying intensities, physical and temporal qualities; (b) extended characterizations at the mol-ecular, cellular and tissue-levels of the radiation?

induced pathologic lesions at the molecular, cellular and tissue levels. The nature of radiation-induced lesion repair, misrepair and/or un-repaired states will be considered

as well; (c) establishment of improved preventive and thera-peutic methods for radiation injury. This includes preventive methods for injuries from external, deeply

penetrating ionizing rays and from externally/internally deposited radionuclides; and (4) identification, characterization, and development of medical strategies to counter

the enhanced health risks associated with infections and chemical intoxication within radiation-exposed personnel.

C. Deliverables
16. Technical reports will be produced on: (a) identification of new biodosimetric options;(b) development of a prototype of a lead candidate biodosimetry assessment

tool; (c) development of improved preventive treatment protocols for hematopoietic tissue injuries; (d) development of improvd medical approaches in preventing

infections and limiting severity of chemical intoxications following ionizing radiation exposures. D. Duration of Technical Team
17. The planned duration of the RTG is 3 years; June 1, 2005 through May 31, 2008.


III. RESOURCES
A. Membership
18. The proposed Task Group will be composed of representatives from member nations of the Human Factors and Medicine (HFM) Panel. The membership is open. The

group will be composed of specialists (civilian and military) in physiology and medicine (emphasis on radiobiology, biochemistry, molecular and cellular biology, and

immunology) and in health physics and nuclear physics (emphasis on spectroscopy, dosimetry, and radiation transport). As needed, further experts may be invited to

join research RTG discussion sessions to provide expertise and advice in specific areas. This RTG would strongly benefit from a multiple disciplinary composition (physicists, biologists, and medical officers) to meld the contributing aspects and recommendations regarding detection of dose, dose-rate, nuclides involved, and
health implications.
19. NATO nations participating (tentative list) include: CA, CZ, FR, GE, HU, NO, PO, SW, TU, UK, and US. An updated list of participating nations on the RTG will be made

by the HFM via liaison reports.
20. The Technical Team Leader will be Dr William F. Blakely, US. The Technical Leader will not serve as a national representative at the same time.
21. The acting pilot nation for the RTG will be the US. RTG delegates will through consensus provide a recommendation to the HFM panel for consideration and

confirmation.
B. National/NATO Resources Required
22. Sufficient resources in terms of personnel, equipment, and supplies are currently available within the research facilities of each of the participating member nations.
23. General administrative guidance and support from NATO/RTA is requested, along with technical and financial assistance in publishing and distributing technical

reports. Financial support for RTG-associated travel, including PfP delegates and RTG consultants is requested as well.
IV. SECURITY CLASSIFICATION LEVEL
24. Up to and including NATO SECRET. Meetings will normally be NATO UNCLASSIFIED.
V. PARTICIPATION BY PARTNER NATIONS
25. Membership of the RTG is open; participation by partner nations is available.
VI. LIAISON
26. Liaison and consultation will take place with the NATO NBC Medical Working Group, and the Radiological Hazard TP 13 Group. The RTG-099 will seek liaison officers

with other relevant NATO Working Groups.

Acknowledgements
Dr. Marek K. Janiak (POL) served as the Referee for The Radiation Bioeffects and Countermeasures RTG-033 and co-chaired subgroup #1. He provided sustained and

critical guidance that focused RTG-033 activities to accomplish its goals. Additional RTG-033 members who served as co-chairs for the four subgroups are shown in the

table below. The subgroup chairs are primarily responsible for establishing the subgroup tasks at onset and the deliverables included in this Final Report. The lists of RTG-033 members as well as contributors to components of this report, who participated and

contributed to the success of RTG-033 tasks, are also included in this report.

RTG-033 subgroups and co-chairs
1. Radiobiology mechanisms and late effects
Co-chairs: Dr. Marek K. Janiak (POL), Dr. Alexandra C. Miller (USA), and Dr. Michael Abend (DEU)
2. Radiation injury assessment and biodosimetry
Co-chairs: Dr. Diana Wilkinson (CAN), Dr. Daniela L. Stricklin (SWE), and Dr. David Holt (GBR)
3. Prophylaxis and therapy
Co-chairs: Dr. Francis J. Herodin (FRA), Dr. Mark H. Whitnall (USA) and COL Patricia Lillis-Hearne (USA)
4. Combined injuries and treatment
Co-chairs Dr. Terry C. Pellmar (USA), Dr. Daniela L. Stricklin (SWE), and Dr. David Holt (GBR)

There were four annual RTG-033 meetings that were instrumental in the conduct of the RTG-033 activities.
The respective host nations, Institutes’ leadership, and local RTG-033 members are to be commended for facilitation of the RTG-033 activities. The local hosts extended numerous travel-related assistance and social activities to the invited meeting attendees, which were very much appreciated.

Date RTG-033 annual meeting location
June 2005 Armed Forces Radiobiology Research Institute (AFRRI), Bethesda, Md. USA
June 2006 Swedish Defense Research Agency (FOI), Umea, Sweden
June 2007 University of Defence, Brno, Czech Republic
June 2008 Centre de Recherches du Service de Sante des Armees, La Tronche, France
LtCol Charles A. Salter provided outstanding service as vice-chair for the first two years of the RTG-033 activities. The RTG-033 chair also extends thanks to several RTG-033 members (Dr. Marek J. Janiak, POL; COL Jan Osterreicher, Czech Republic; and COL Patricia Lillis-Hearne, USA) who provided liaison and consultation to NATO HFM Panel and CBRN Medical Working Group. In addition, the excellent editorial, publishing, and graphic support services by the Armed Forces Radiobiology Research Institute’s dedicated support staff are gratefully acknowledged.

William F. Blakely (USA)
RTG-033 Chair

RTG-033 Group Members

Canada
Dr. Diana Wilkinson
Defence R&D Canada
3701 Carling Avenue
Ottawa, Ontario K1A 0Z4 Canada
Tel: 1 613 998-5995
Fax: 1 613 998-4560
diana.wilkinson@drdc-rddc.gc.ca

Dr. Ian D. Torrie
Canadian Forces
1745 Alta Vista Drive
Ottawa, Ontario K1A 0K6 Canada
Tel: 1 613 945-6600
Fax: 1 613 945-6668
Torrie.ID@forces.gc.ca

Dr. Slavica Vlahovich
CF Health Services Group
Department of Defense
116-1745 Alta Vista Drive
Ottawa, Ontario K1A 0K6 Canada
Tel: 1 613 945-6600
Fax: 1 613 945-6668
Vlahovich.S1@forces.gc.ca

Dr. Edward J. Waller
University of Ontario Institute of Technology
2000 Simcoe Street North
Oshawa, Ontario L1H 7K4 Canada
Tel: 1 905 721-3111
Fax: 1 905 721-3370
ed.waller@uoit.ca

Czech Republic
Dr. Jan Osterreicher
University of Defence
Purkyne Mil Med Faculty Trebesska 1575
Hradec, Czech 500 02
Tel: 004 973 25 32 19
Fax: 004 49 551 3018
J.Osterreicher@seznam.cz


France
Dr. Francis Jean Herodin
Centre de Recherches du Service
de Sante des Armees (CRSSA)
24, avenue des Maquis du Gresivaudan
La Tronche cedex, Rhone-Alpes 38702 France
Tel: 33 47663 6933
Fax: 33 47663 6922
fherodin@crssa.net

Dr. Didier Clarencon
Centre de Recherches du Service
de Sante des Armees (CRSSA)
24, avenue des Maquis du Gresivaudan
La Tronche cedex, Rhone-Alpes 38702 France
Tel: 33 47663 6926
Fax: 33 47663 6922
didierclarencon@crssa.net

Germany
Dr. Michael Abend
Bundeswehr Institute of Radiobiology
Federal Armed Forces Medical Academy
Neuherbergstr. 11
Munich, Bavaria 80937
Tel: 049 89 3168-2280
Fax: 049 89 3168-2255
michaelabend@bundeswehr.org

Great Britain
Lt Col David Charles Bates
Defence Medical Services Department
7/G/17 Main Building
Whitehall, London SW1A 2HB UK
Tel: ++44 (0)207 807 0386
Fax: ++44 (0)207 218 1447
david-bates762@mod.uk

Dr. David C.B. Holt
Institute of Naval Medicine
Crescent Road Alverstoke
Gosport, Hampshire PO12 2DL UK
Tel: +44 23 8085
Fax: +44 23 48237
smorm@inm.mod.uk

Dr. David C. Lloyd
Health Protection Agency
Chilton, Didcot, Oxfordshire
OX11 0RQ United Kingdom
Tel: 44 1235 822700
Fax: 44 1235 833891
david.lloyd@hpa.org.uk

Mr. Alan Hodgson
Health Protection Agency
Building 566, Chilton
Didcot, Oxfordshire
OX11 0RQ, UK
Tel: 44 01235 822653
Fax: 44 01235 833891
alan.hodgson@hpa-rp.org.uk

Italy
Dr. Marco Durante
University Federico II
Monte S. Angelo, Via Cintia
Napoli, Italy 80131
Tel: 39 081 676440
Fax: 39 081 676346
durante@na.infn.it

Netherlands
Mr. Maarten Huikeshoven
CEMG, Ministerie van Defensie
Noodweg 15, Hilversum
Utrecht, Netherlands
Tel: 31 35 577-4534
Fax: 31 35 577-4530
mj.huikeshoven@mindef.nl

Dr. Ad S de Koning
Ministry of Defense, CEMG
PO Box 155
Loosdrecht, Noord Holland
1230 AD Netherlands
Tel: 31 (0)35 5774533
Fax: 31 (0)35 5774530
As.d.koning@mindef.nl

Norway
Dr. Alicja Jaworska
Norwegian Radiation Protection Authority
POB 55
Oesteraas Akershus 1361 Norway
Tel: 47-67162500
Fax: 47-67147407
Alicja.Jaworska@nrpa.no

Poland
Dr. Marek K. Janiak
Military Institute of Hygiene & Epidemiology
4 Kozielska St.
01-163Warsaw, Poland
Tel: + 48 22 681 8518
Fax: + 48 22 810 4391
mjaniak@wihe.waw.pl

Sweden
Dr. Daniela L. Stricklin
FOI, Swedish Defense Research Agency
Cementvagen 20
Umea, Vasterbotten 90628 Sweden
Tel: 46 90 106746
Fax: 46 90 106803
Current address:
Applied Research Associates
801 N. Quincy Street, Suite 600
Arlington, VA 22203
Tel: 703-816-8886
Fax: 701-816-8861
dstricklin@ara.com

United States
COL Patricia K. Lillis-Hearne
AFRRI
8901 Wisconsin Avenue, Bldg 42
Bethesda, MD 20889-5603 USA
Tel: 1 301 295-1210
Fax: 1 301 295-4967

Dr. Terry C. Pellmar
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1 301 295-1211
Fax: 1 301 295-4967
pellmar@afrri.usuhs.mil
Current email address:
tpellmar@yahoo.com

Dr. William F. Blakely
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1 301 295-0484
Fax: 1 301 295-1863
blakely@afrri.usuhs.mil

Dr. Mark H. Whitnall
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1 301 295-9262
Fax: 1-301 295-6503
Whitnall@afrri.usuhs.mil

Dr. Alexandra C. Miller
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-
Fax: 1-301-295
Miller@afrri.usuhs.mil

Dr. Vitaly Nagy
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1 301 295-0378
Fax: 1 301 295-0375
nagy@afrri.usuhs.mil

LTC John Cuellar
Uniformed Services University
of the Health Sciences
4301 Jones Bridge Road
Bethesda, MD 20814-4799 USA
Tel: 1 301 295-3309
Fax: 1 301 295-3320
john.cuellar@us.army.mil

Mr. Tommy Morris
TATRC
MRMC, Fort Detrick
Building 1054
504 Scott Street
Fort Detrick, MD 21702-5012 USA
Tel: 1 301 619-7938
Fax: 1 301 619-7968
morris@tatrc.org

Mr. Jason Dunavant
Command Surgeon’s Office
HQ Army Materiel Command
ATTN: AMCPE-SG-R
9301 Chapek Road
Fort Belvoir, VA 22060 USA
Tel: 1 703 806-8714
Fax: 1 703 806-8859
Jason.dunavant@us.army.mil
Canada
Dr. Diana Wilkinson
Defence R&D Canada
3701 Carling Avenue
Ottawa, Ontario K1A 0Z4 Canada
Tel: 1 613 998-5995
Fax: 1 613 998-4560
diana.wilkinson@drdc-rddc.gc.ca

Dr. Edward J. Waller
University of Ontario Institute of Technology
2000 Simcoe Street North
Oshawa, Ontario L1H 7K4 Canada
Tel: 1 905 721-8668 x2609
Fax: 1 905 721-3046
ed.waller@uoit.ca

France
Dr. Francis Jean Herodin
Centre de Recherches du Service
de Sante des Armees (CRSSA)
24, avenue des Maquis du Gresivaudan
La Tronche cedex, Rhone-Alpes 38702 France
Tel: 33 47663 6933
Fax: 33 47663 6922
fherodin@crssa.net

Dr. Jean-Francois Mayol
Centre de Recherches du Service
de Sante des Armees (CRSSA)
24, avenue des Maquis du Gresivaudan
La Tronche cedex, Rhone-Alpes 38702 France
Tel: 33 476 639754
Fax: 33 476 636922
mayol@crssa.net

Dr. Michel Drouet
Centre de Recherches du Service
de Sante des Armees (CRSSA)
24, avenue des Maquis du Gresivaudan
La Tronche cedex, Rhone-Alpes 38702 France
Tel: 33 476 636931
Fax: 33 476 636922
micheldrouet@crssa.net


Germany
Dr. Michael Abend
Bundeswehr Institute of Radiobiology
Federal Armed Forces Medical Academy
Neuherbergstr. 11
Munich, Bavaria 80937
Tel: 049 89 3168-2280
Fax: 049 89 3168-2255
michaelabend@bundeswehr.org

Dr. Matthias Port
Clinic for Hematology, Hemostaseology, Oncology
and Stell Cell Transplantation
Hannover Medical School
Hannover, Germany
Tel: +49-511-532-3019
Fax: +49-511-532-8049
Matthias.port@web.de

Dr. Armin Riecke
Bundeswehr Institute of Radiobiology
Federal Armed Forces Medical Academy
Neuherbergstr. 11
Munich, Bavaria 80937
Tel: 049 89 3168-2280
Fax: 049 89 3168-2255
ArminRiecke@bundeswehr.org

Dr. Christian G. Ruf
Bundeswehr Institute of Radiobiology
Federal Armed Forces Medical Academy
Neuherbergstr. 11
Munich, Bavaria 80937
Tel: 049 89 3168-2280
Fax: 049 89 3168-2255
christruf@gmx.de

Great Britain
Dr. David C.B. Holt
Institute of Naval Medicine
Crescent Road Alverstoke
Gosport, Hampshire PO12 2DL UK
Tel: +44 23 8085
Fax: +44 23 48237
smorm@inm.mod.uk

Dr Kai Rothkamm
Health Protection Agency
Centre for Radiation, Chemical and Environmental
Hazards Fermi Avenue Chilton, Didcot, Oxon
OX11 0RQ
United Kingdom
Tel: +44 (0)1235 822700
Fax: +44 (0)1235 833891
Email: kai.rothkamm@hpa.org.uk

Norway
Dr. Alicja Jaworska
Norwegian Radiation Protection Authority
POB 55
Oesteraas Akershus 1361 Norway
Tel: 47-67162500
Fax: 47-67147407
Alicja.Jaworska@nrpa.no

Poland
Dr. Marek K. Janiak
Military Institute of Hygiene & Epidemiology
4 Kozielska St.
01-163Warsaw, Poland
Tel: + 48 22 681 8518
Fax: + 48 22 810 4391
mjaniak@wihe.waw.pl

Dr. Ewa M. Nowosielska
Military Institute of Hygiene & Epidemiology
4 Kozielska St.
01-163Warsaw, Poland
Tel: + 48 22 681 6135
Fax: + 48 22 810 4391
ewan14@wp.pl

Dr. Aneta Cheda
Military Institute of Hygiene & Epidemiology
4 Kozielska St.
01-163 Warsaw, Poland
Tel: + 48 22 681 6135
Fax: + 48 22 810 4391
acheda@wp.pl

Dr. Jolanta Wrembel-Wargocka
Military Institute of Hygiene & Epidemiology
4 Kozielska St.
01-163 Warsaw, Poland
Tel: + 48 22 681 6135
Fax: + 48 22 810 4391
wwkasia@poczta.fm

Sweden
Dr. Daniela L. Stricklin
FOI, Swedish Defense Research Agency
Cementvagen 20
Umea, Vasterbotten 90628 Sweden
Tel: 46 90 106746
Fax: 46 90 106803
Current address:
Applied Research Associates
801 N. Quincy Street, Suite 600
Arlington, VA 22203
Tel: 703-816-8886
Fax: 701-816-8861
dstricklin@ara.com

United States
COL Patricia K. Lillis-Hearne
AFRRI
8901 Wisconsin Avenue, Bldg 42
Bethesda, MD 20889-5603 USA
Tel: 1 301 295-1210
Fax: 1 301 295-4967
smithy@afrri.usuhs.mil

Dr. Terry C. Pellmar
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1 301 295-1211
Fax: 1 301 295-4967
pellmar@afrri.usuhs.mil
Current email address:
tpellmar@yahoo.com

Dr. William F. Blakely
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1 301 295-0484
Fax: 1 301 295-1863
blakely@afrri.usuhs.mil
Dr. Mark H. Whitnall
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1 301 295-9262
Fax: 1 301 295-6503
Whitnall@afrri.usuhs.mil

Dr. Alexandra C. Miller
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-9232
Fax: 1-301-295-6503
Miller@afrri.usuhs.mil

Dr. Natalia I. Ossetrova
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-1619
Fax: 1-301-295-0313
Ossetrova@afrri.usuhs.mil

Dr. Gregory L. King
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-2861
Fax: 1-301-295-0292
King@afrri.usuhs.mil

Dr. Kyle Millage
Applied Research Associates, Inc.
801 N. Quincy St., Suite 600
Arlington, VA, 22203, USA
Tel: 1-908.782.8204
Fax: 1-908.782.7526
Kyle.millage@ara.com

Dr. Steven G. Homann
Lawrence Livermore National Laboratory
7000 East Ave.
Livermore, CA, 94550, USA
Tel:1-925-423-4962
Fax: NA
shomann@llnl.gov

Dr. Gayle Sugiyama
Lawrence Livermore National Laboratory
7000 East Ave.
Livermore, CA, 94550, USA
Tel: 1-929-422-7266
Fax: 1-925-422-6388
sugiyama@llnl.gov

Dr. John S. Nasstrum
Lawrence Livermore National Laboratory
7000 East Ave.
Livermore, CA, 94550, USA
Tel: 1-925 423-6738
Fax: 1-925 423-4527
jnasstrom@llnl.gov

Dr. Jeffrey B. Nemhauser
c/o Radiation Studies Branch
Center for Disease Control and Prevention
4770 Buford Highway, NE
Atlanta, GA 30341-3717 USA
Tel: 1-770.488.3658
Fax: 1-770.488.1539
jnemhauser@cdc.gov

Dr. Brooke R. Buddemeier
Lawrence Livermore National Laboratory
7000 East Ave.
Livermore, CA, 94550, USA
Tel: 1-925-423-2627
Fax: 1-925-422-9343
buddemeier1@llnl.gov

Uma Subramanian
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-1615
Fax: 1-301-295-6857
Subramanian@afrri.usuhs.mil

Katya Krasnopolsky
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-0306
Fax: 1-301-295-0313
Krasnopolsky@afrri.usuhs.mil
Patrick Martin
PSC 111Box 3B
APO, AE 09454
Tel: 443.602.9715 (MD Internet phone)
patrick@readyallrow.com

Deena S. Disraelly
Strategy, Forces & Resources Division
Institute for Defense Analyses
4850 Mark Center Drive
Alexandria, VA 22311-1882
Phone: 1-703-845-6685
FAX: 1-703-845-2255
ddisrael@ida.org

Carl A. Curling, Sc.D.
Strategy, Forces & Resources Division
Institute for Defense Analyses
4850 Mark Center Drive
Alexandria, VA 22311-1882
Tel: 1-703-578-2814
FAX: 1-703-575-4682
ccurling@ida.org

David J. Sandgren
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-5128
Fax: 1-301-319-0311
Sandgren@afrri.usuhs.mil

Ira H. Levine
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603
Tel: 1-301-295-9130
Fax: 1-301-295-6503
Levine@afrri.usuhs.mil

Dr. Venkataraman Srinivasan
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-5826
Fax: 1-301-295-6503
srinivasan@afrri.usuhs.mil

Dr. Michael R. Landauer
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-5606
Fax: 1-301-295-6503
landauer@afrri.usuhs.mil

Dr. Vijay K. Singh
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-2347
Fax: 1-301-295-6503
singh@afrri.usuhs.mil

Dr. Mang Xiao
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-2597
Fax: 1-301-295-6503
xiao@afrri.usuhs.mil

Dr. Marcy B. Grace
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Current address:
Chemical, Radiological & Nuclear Medical
Countermeasures
Biomedical Advanced Research & Development
Authority
330 Independence Avenue, SW
Room 644G
Washington, DC 20201 USA
Tel: 1-202-205-9802
marcybgrace@yahoo.com

Dr. K. Sree Kumar
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-0083
Fax: 1-301-295-6503
kumar@afrri.usuhs.mil




Dr. Martin Hauer-Jensen
Arkansas Cancer Research Center, Suite 241
4301 West Markham, Slot 725
Little Rock, AR 72205
Tel: 1-501-686-7912
Fax: 1-501-421-0022
mhjensen@uams.edu

Dr. Sanchita P. Ghosh
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-1945
Fax: 1-301-295-6503
ghosh@afrri.usuhs.mil

Dr. Steven R. Mog
FDA ? CFSAN
Office of Food Additive Safety
5100 Paint Branch Parkway (HFS ? 205)
College Park, MD 20740 USA
Tel: 1-301-436-1190
Fax: 1-301-436-2973
steven.mog@fda.hhs.gov

Dr. Cheng-Min Chang
AFRRI
8901 Wisconsin Avenue
Bethesda, MD 20889-5603 USA
Tel: 1-301-295-5544
Fax: 1-301-295-6503
chang@afrri.usuhs.mil

Dr. Tzu-Cheg Kao
Uniformed Services University
4301 Jones Bridge Road
Bethesda, MD 20814 USA
Tel: 1-301-295-9756
Fax: 1-301-295-1854
tkao@usuhs.mil


NATO Human Factors and Medicine Panel (HFM)-099
Research Task Group (RTG)-033 Activity
Radiation Bioeffects and Countermeasures

Chapter 1
Summary of Activities
Dr. William F. Blakely (USA)
Chair
1.0 INTRODUCTION
The objective of this Research Task Group was to develop the scientific basis for new and improved methods to prevent, assess, treat, and manage casualties and long-term health effects associated with ionizing radia-tion exposure from evolving threats in military operations. RTG-033 was organized into four subgroups to focus on the delivery of radiobiology-related products (Figure 1). Participation was by NATO member (10) and partner (1) nations‘ representatives. Membership excluded epresentatives from Mediterranean Dialogue
(MD) nations.

Figure 1: NATO HMF-099/RTG-033 organization and products
RTG-033 meetings were held annually and hosted by various nations‘ representatives (Table 1) during the group‘s tenure.

Program objectives were developed over a period of four years; the first meeting was held in the USA in 2005 (see Appendix for agenda). At this inaugural meeting the Working Group agreed to explore new and improved methods to prevent, assess, treat, and manage casualties and long-term health effects associated with ionizing radiation exposure from evolving threats in military operations. Four subgroups were established and each was tasked with the responsibility of identifying contributing RSG members and developing a Technical Activity Program (TAP) that will address specific aims, tasks and a schedule for deliverable milestones.
The second meeting was held in Sweden in 2006 (see Appendix for agenda). At this meeting each subgroup had the opportunity to review advancements made toward specific goals set in the first meeting, re-evaluate members‘ contributions to each area of interest, identify milestones and establish timelines for project achievements.
The third meeting was in June 2007 and was hosted by members from the Czech Republic (see Appendix for agenda). This meeting provided an interactive opportunity for participants to further discuss the specific activities agreed upon in the TAP and initiate discussions for future Technology Demonstration activity.
The fourth and final meeting was held in France June 2008 (see Appendix for agenda). It enabled organi-zation and integration of the latest scientific advancements into a final report. At this meeting a representative from Canada invited RTG-033 members to participate in planning and execution of a biodosimetry technology demonstration activity under the Defence Against Terrorism (DAT) program funding, scheduled to occur after the completion of RTG-033 activities.
Through this four-year interactive and collaborative Working Group, Subgroup 2 has been able to bring to the table international expertise and progress toward testing deployable capabilities. The outcome of this subgroup will further the deployment capabilities and improve methods to prevent, assess, treat, and manage casualties and long-term health effects associated with ionizing radiation exposure.

The co-chairs for the four subgroups established their specific aims and tasks as described below.
Sugroup 1?Radiobiology Mechanisms and Late Effects The group's specific aims/tasks include:
a. Develop and use models to study late effects of radiation [USA].
b. Examine low level radiation-induced effects and their mechanisms [POL].
c. Improve understanding of the acute radiation syndrome (ARS) [FRA, GER, POL, USA].
d. Identify potential markers of radiation injury [GER, POL, USA].
Subgroup 2?Radiation Injury Assessment and Biodosimetry
The group's specific aims/tasks were:
Table 1: Annual meetings of RTG-033
Date Meeting location
June 2005 Armed Forces Radiobiology Research Institute (AFRRI), Bethesda, Md. USA
June 2006 Swedish Defense Research Agency (FOI), Umea, Sweden
June 2007 University of Defence, Brno, Czech Republic
June 2008 Centre de Recherches du Service de Sante des Armees, La Tronche, France


a. Develop an evaluation tool that will guide the selection, advancement, implementation and vali-dation of complementing biodosimetry assays [GBR].
b. Develop and validate lead candidate multi-parameter biodosimetry tools [CAN, CZE, GER, FRA, GBR, NOR, POL, SWE, USA].
c. Evaluate software applications designed to provide radiation injury assessment, biological dosimetry information and guidance to the first-responder and first-receiver communities [CAN, CZE, GER, FRA, GBR, SWE, USA].
d. Develop injury assessment and biological dosimetry guides and documents [GBR, USA].
e. Develop a proposal for Advanced Demonstration Technology by the final RTG-099 meeting [CAN, CZE, GER, FRA, GBR, POL, SWE, USA]. Subgroup 3?Prophylaxis and Therapy
The group's specific aims/tasks included:
a. Identify and develop next-generation prophylaxis and therapeutic agents [FRA, POL, USA].
b. Preclinical testing of candidate drugs and subsequent clinical validation of efficacies [FRA, POL, USA].
c. Advanced development of candidate drugs (large animals, clinical safety and PK) [FRA, USA].
Subgroup 4 Combined Injuries and Treatment The group‘s specific tasks were:
a. Assessment of combined injury: Define progress; identify gaps and directions including biomarkers and animal effects [USA, SWE, GBR, CAN].
b. Countermeasures: Define status of approaches; identify gaps [USA, GBR, NOR, CAN].
c. Modeling of combined effects starting with blast and burn [USA, GBR].

Designated members of the RTG-033 annually provided liaison and consultation with NATO‘s HFM Panel and CBRN Medical Working Group during the tenure of RTG-033 activities.

Subsequent sections present the final reports for the respective subgroups.


1.1 SubGroup #1?Radiobiology Mechanisms and Late Effects:
Summary of Activities

M.K. Janiak (POL), A. C. Miller (USA), and M. Abend (GER)
Subgroup #1 Co-Chairs

The growth of criminal (including terrorist) activities as well as dissemination of nuclear materials and technologies has recently increased the probability of both radiological and nuclear threats. Such threats are primarily associated with low levels (dirty bomb) and late effects of radiation, but exposures to higher doses leading to the acute radiation syndrome (ARS) also are possible (nuclear explosions, radiation accidents, etc.). In view of this, the purpose of this subgroup was to: 1. develop and use models to study late effects of radiation, 2. examine low level radiation-induced effects and their mechanisms, 3. identify poten-tial markers of radiation injury and its consequences (i.e., sporadic thyroid cancer), and 4. improve under-standing of the ARS syndrome. Subgroup 1‘s Final Reports include characterization at the molecular,
cellular, and tissue levels of radiation-induced effects. Four program objectives were addressed during the tenure of activities with presentations on these topics at each of the four annual meetings; see Appendix for annual meeting agendas. Task items 1 through 4 are summarized below:
1. To study the carcinogenic effects of external radiation or internal emitters such as depleted uranium (DU), we have developed a rodent leukemia model (USA). External whole-body exposure to radiation or chronic exposure to embedded DU pellets has been shown to be leukemogenic. This model allows us to identify and monitor a range of late-effect biomarkers such as chromosomal alterations, hematological parameters, and molecular markers. Secondly, this model enables us to test promising medical countermeasures and determine whether they are beneficial in the prevention of radiation-induced leukemia. Recent studies demonstrating radiation and DU transgenerational effects in un-exposed offspring were conducted using the ―Big Blue? transgenic mouse model. This system allows us to assess genomic instability in unexposed offspring and in the exposed parent. Data demonstrate that chronic and high-dosage DU-exposure of fathers induced a significant increase in the mutation fre-quency in both paternal testes and offspring bone marrow. This model allows us to study trans-generational effects including offspring susceptibility to cancer. In vitro models also have been developed at AFRRI to assess radiobiology basic mechanisms. Neoplastic transformation studies are used to assess carcinogenicity and to screen promising radioprotectors. The hypoxanthine guanine phos-phoribosyl transferase (hprt) mutation assay not only has been used to evaluate mutagenicity but has shown promise as an alpha particle exposure biomarker. In addition, the relationship between radiation ―bystanders? and adaptive response are being investigated using immortalized human cells.
2. Tumor growth and radiation-boosted anti-neoplastic immune reactions have been assessed using a murine model (POL). It has been demonstrated that both single and fractionated exposures of mice to 0.1 or 0.2 Gy X-rays inhibit the development of artificial metastases and stimulate cytotoxic activities of splenocytes enriched for natural killer cells (NK splenocytes) and activated peritoneal macrophages (Mφ). This effect was associated with the significantly stimulated Mφ- and NK splenocytes-mediated cytolysis of susceptible tumor targets and, in the case of Mφ, the elevated production of NO. The enhanced cytolytic activity of NK splenocytes was mediated by perforin and the Fas receptor ligand. Both single and fractionated irradiation of mice with 0.1, 0.2, or 1.0 Gy X-rays stimulated secretion of IFN-γ and IL-2 by
the NK splenocytes as well as IL-1β, IL-12, TNF-α and Mφ. Single exposures of isolated NK splenocytes and Mφ to 0.1 or 0.2 Gy X-rays neither boosted the cytotoxic activities of these cells nor enhanced their rate of apoptosis. In conclusion, the data indicate that suppression of the development of pulmonary tumor colonies by single or fractionated irradiations of mice with the two low doses of X-rays may, at least in part, be due to stimulation of natural defense reactions mediated by NK lymphocytes and/or cytotoxic macrophages. Also, results suggest that boosting of the cytolytic functions of these anti-tumor cells by low-level X-ray exposures relies on interaction with other environmental cells and factors that occur in in vivo but not in vitro conditions.
3a. In general, the cause of a developing tumor and in particular the causal link to ionizing radiation cannot be made. However, due to possible compensation requests of exposed individuals it is desirable to be able to do that. In a previous study we found differences in gene expression of several genes that made it possible to completely distinguish radiation associated (post-Chernobyl) from sporadic thyroid cancer (GER).
3b. Cell death mechanisms that predominate at higher doses are examined on the gene-expression level (GER)?several in vitro and in vivo models were established. The dose-response relationship of single genes and signal transduction pathways was examined. These pathways code biological processes such as cell proliferation, different modes of cell death, and DNA-repair. An RTQ-PCR based platform was created (CLARCC array).
3c. The acute radiation syndrome or sickness (ARS) is characterized by dose- and time-dependent expression of various organ and tissue specific sub-syndromes (i.e., hematopoietic, gastrointestinal, and cardiovascular). The dynamic time-course of these clinical signs and symptoms form the basis for a standardized biological-based scoring system to quantify the severity levels of the various sub-syndromes. Recently the use of molecular or metabolic biomarkers, many detected in blood plasma or
serum, have been advocated as additional diagnostic indexes to aid early triage and to monitor the efficacy of medical treatment. Subgroup#1 Final Report also includes a review of the current literature on this topic (USA, GER). Within this scope of items future experiments will include: a) development of pharmacological counter-
measures for internal and external radiation-induced leukemia and assessment of diagnostic leukemia bio-markers; b) further elucidation of the kinetics and mechanisms of anti-tumor functions of NK cells and macrophages stimulated by both single and fractionated low-level irradiations and establishment of the plausible threshold doses for these effects; c) extending the sporadic thyroid cancer-gene expression study by increasing the number of thyroid cancers (TC) and improving the experimental design in order to determine the usefulness of gene expression for discriminating radiation-associated from sporadic TC; d) continue measurements in a set of genes that, irrespective of radiation dose, the cell model and the form of cell death, appear to be altered after irradiation; it is also planned to extend these gene expression studies to include use of samples from thyroid cancer patients treated with radionuclides, and e) evaluation of a multiple organ biomarker approach to compliment the conventional ARS diagnostic methodology in a nonhuman primate radiation model.
1.2 RTG 033 Subgroup 2: Radiation Injury Assessment and Biodosimetry: Summary of Activities

Diana Wilkinson (CAN), David C.B. Holt (GBR), Daniela L. Stricklin (SWE)
Subgroup 2 Co-Chairs

Medical and scientific communities are well aware of the challenges to be encountered in management of radiological/nuclear mass-casualty disasters. Available medical resources will need to be controlled strin-gently and directed towards optimal outcomes. In recognition of these requirements, NATO Research Task Group HFM-099/RTG-033 undertook the development of new and improved methods/tools to assist the medical community to prevent, assess, treat, and manage casualties and long-term health effects associated with ionizing radiation exposure. The primary goal of all emergency response communities is to prevent or minimize the likelihood a radiological/nuclear event occurrence; however, consequence management procedures still require establishment and testing. Subgroup #2 undertook the tasking to develop improved methods for casualty assessment. Five specific and interrelated tasks were identified by the subgroup as targets for advanced research. All Member States have agreed to develop an evaluation tool, based on the Technology Readiness Level (TRL) assessment, which would guide the selection, advancement,
implementation and validation of available and upcoming biodosimetry assays. Presently there is no single biodosimetry assay that is uniquely suitable for all scenarios. The selection of the most appropriate assay will depend on the unique circumstances, the number of casualties that need to be assessed, and available resources. An ideal dosimeter or more likely a combination of the most suitable dosimeters needs to be identified. The outcome of this process will result in a tool that will assist casualty management controllers in their selection of the most appropriate and relevant biodosimetry methods for the particular scenario. The proposed tool, based on the TRL evaluation process, also will offer guidance to the Member State‘s research programs in order to identify the lead candidate multi-parameter biodosimetry tools for advanced development and validation. In parallel, evolution of software applications designed to provide radiation injury assessment, biological dosimetry information and guidance to the first-responder and first-receiver communities will offer a range of field-deployable, emergency-response-assistance tools developed by the Member States. Subgroup #2 reports are conveyed reports (Chapters 6?8) and include a comprehensive summary of the subgroup‘s products (Chapter 6) along with reports on biodosimetry recommendations for mass-casualty incidents (Chapter 7) and on-going effects to automate the processing of cytogenetic analysis for dose assessment (Chapter 8). With the knowledge gained through these activities, Subgroup #2 members are in a position to offer a pool of international experts capable of providing the latest advice in the development of injury assessment and biological dosimetry guides and documents. The final task of this subgroup, after the completion of RTG-033 activities, is to participate in a NATO exercise through the Advanced Demonstrations Technology process to demonstrate the latest advancements in radiation-injury
assessment and biodosimetry. This process will enable future opportunities for field testing of alternate methods that can build upon present capabilities.
1.3 RTG 033 Subgroup 3: Prophylaxis and Therapy: Summary of Activities  

  拍手はせず、拍手一覧を見る

コメント
 
01. SaveChild厨 2012年7月12日 01:00:48 : hfhZnjAARar8w : o2YvAF0OXo

Francis J. Herodin (FRA), Mark H. Whitnall (USA), and Patricia K. Lillis-Hearne (USA)
Subgroup #3 Co-Chairs

Although the Acute Radiation Syndrome (ARS) has been well documented at the clinical level, and mech-anistic information is accumulating rapidly, successful prophylaxis and treatment for ARS is problematic.
There is a pressing need to develop radiation countermeasures that can be used both in the clinic and outside the clinic in operational and mass-casualty scenarios. Subgroup 3 of NATO HFM-099/RTG-033 was estab-lished to direct research on basic and applied research leading to development of safe and effective radiation countermeasures and to report findings to the RTG.
At the inaugural meeting of the RTG in the US in June 2005, the characteristics of a successful counter-measure were discussed in the context of likely scenarios. Strategies for discovering and developing countermeasures candidates were considered and the compounds under development were listed. High-lighted were findings on 5-androstenediol (5-AED), the tocols, genistein, and cytokines. Keynote talks included "Current Status of Treatment of Radiation Injury in the United States" by COL David G. Jarrett, US, and "Current STANAGS and Concept/Capability Gaps?Prioritized Needs as Identified by the NATO
NBC Medical WG by Wing Commander Victor J. Wallace, UK. It was agreed to form Subgroup 3 to extend these activities, with co-chairs from the US and France.
At the second meeting in Sweden in June 2006, the current status of radiation countermeasure research was updated, with added material on mechanisms of action of the most promising candidates, chelation of internalized radionuclides, and pegylated cytokines. The two keynote presentations directly relevant to Sub-group 3‘s aims were "Medical emergency preparedness for activities connected to remediation of Adreeva Bay?Norwegian and Russian Federation regulatory cooperation between authorities and experts," by Dr. Alicja Jaworska, Norway, and "Mitigating the clinical effects of ionising radiation: An update on perceived capability and research gaps from the NATO NBC Medical Working Group," by Dr. David C. Bates, UK.
The third meeting in the Czech Republic in June 2007 presented advances in studies on the candidates discussed at previous meetings, and also included a presentation on cell and gene therapy.
The fourth and final meeting was held in France in June 2008. Two keynote lectures related to the subgroup‘s goals: "Medical management of irradiated victims in nuclear/biological accidents or mass-casualty acts of
terrorism," by Dr. T. De Revel, and "State of art of the medical management of radiological burns," by Dr. Herve Lebever, both of France. In addition to updates on countermeasures in development, there was an added
emphasis on responses to radiation dispersal devices, with a talk by Dr. Daniela Stricklin on that subject.
It is clear from the research and development efforts discussed in Subgroup 3 that promising countermeasure candidates exist at the preclinical stage. Development programs are moving into trials in large animal models as
well as clinical safety trials, with the appropriate interactions with regulatory agencies taking place to facilitate eventual approval of these countermeasures as human pretreatments and therapies. No countermeasures for
ARS have been approved for human use in a radiation disaster scenario. The most progress has been made in terms of mitigating the consequences of radiation doses corresponding to the ―hematopoietic syndrome,?
although it is recognized that interactions between multiple organ systems are involved. Efforts are underway to develop countermeasures appropriate for higher radiation doses causing the ―gastrointestinal syndrome.?
1.4 RTG 033 Subgroup 4: Combined Injuries and Treatment: Summary of Activities

Terry C. Pellmar (USA), Daniela L. Stricklin (SWE), and David C.B. Holt (GBR) Subgroup #4 Co-Chairs

Combined injuries of radiation plus trauma or other exposures are likely to be more deadly than any injury or exposure alone. Clinical, animal, and cellular studies suggest synergistic effects. To be fully prepared for a nuclear or radiological event, it will be important to understand the potential interactions, medical con-sequences, and treatment options when combined effects are encountered. In its inaugural meeting, NATO
HFM-099/RTG-033 established a subgroup to address the problem of combined injury. At the first meeting, delegates from the US, UK, SW agreed to co-chair a group to consider the state of the research, status of casualty prediction models as they relate to consideration of combined injuries, and areas of potential labor-atory collaboration.
At the first meeting (June 2005; agenda), the US presented an overview of the effects of radiation in combination with trauma, infectious disease, or chemical exposures. Another presentation addressed the issues associated with antimicrobials in the management of post-irradiation infection. Approaches to dealing with wounds from depleted uranium fragments were discussed in two talks, one providing a synopsis of the status of health concerns about military use of depleted uranium and surrogate metals in munitions, and the other on the development of a colorimetric test for uranium. At this meeting the task group outlined its goals for the three years and began the process of defining the critical aspects of this research effort.
Discussions at the second meeting (June 2006; agenda) centered on biomarkers for combined injuries. The delegates from Sweden discussed concepts regarding the relevance of biomarkers of chemical-induced injury. The delegates from France spoke on their studies of responses to combined injuries in nonhuman primates and relevant neuro-immune biomarkers of prognosis. Discussions at this meeting raised concerns regarding exposures to chemicals in the environment following a radiological or nuclear event. As a result, the focus expanded from only co-exposures of radiation and battlefield trauma or WMD to likely exposures to endemic toxicants, pathogens, and industrial chemicals.
Presentations at the third meeting (June 2007; agenda) addressed the status of ountermeasures for combined injury in radiation threat environment and mathematical modeling of radiation health effects, current state and future plans. These resentations clearly revealed some of the gaps in the field. Few radiation counter-measures had been tested with combined injuries and some of those that were produced nexpected results.
The modeling of combined injury was still in its early stages. It was clear from iscussions that additional experiments were needed to feed into the models and that the models could provide some interesting predictions that should be tested in the laboratory.
At the fourth and final meeting (June 2008; agenda) the presentations focused on modeling efforts. One talk provided an update on mathematical modeling of radiation health effects. Another presentation reviewed the
models of human response to nuclear effects as a function of systems, signs, and symptoms that would be used for operational purposes.
Discussions over the lifetime of this panel have raised awareness of the concerns about combined injuries and stimulated new interactions and collaborative efforts among the NATO members. Countries have sought to contribute by building on their own strengths. Animal research, where it is possible, has contributed to understanding combined injuries in relationship to systemic effects. Cellular research has pointed out new areas of concern. Incorporation of the data into models have benefited from stimulating intellectual discussions among all participating colleagues regarding anticipated scenarios and their consequences. From the interactions of the task group, the efforts on combined injury have evolved into a richer and more mature area of investigation.
Subgroup 4 of NATO HFM-099/RTG-033 has written a final report that summarizes its discussions and interactions. The final report describes 1) the likelihood of combined injuries; 2) the complexities of interactions of radiation with trauma, infectious agents, and chemical agents; 3) the capability of biomarkers of individual susceptibility to provide new diagnostic tools; and 4) models of the human response to combined injury that can guide future studies to define mechanisms, assess outcomes and develop countermeasures.

RTG 033 Subgroup 1: Radiobiology Mechanisms and Late Effects

Chapter 2
Development of Models to Study Radiation-Induced Late Effects
Alexandra C. Miller
Armed Forces Radiobiology Research Institute Uniformed Services University
8901 Wisconsin Avenue, Building 42 Bethesda, MD 20889-5603 USA

ABSTRACT

Military and NATO personnel can potentially be exposed to external and internal radiation during military operations. Radiation exposure can cause late effects like cancer, leukemia, genetic effects, and possible effects to offspring. A multi-parametric in vitro and in vivo approach has shown effectiveness in providing information regarding radiation induced carcinogenicity, promising radiation countermeasures, and trans-generational radiation effects. These in vitro and in vivo models can be used evaluate the specific radiation exposure scenarios experienced by military and NATO personnel.
1.0 INTRODUCTION
Military and NATO personnel can potentially be exposed to external and internal radiation during military operations. The external radiation can occur via a nuclear weapons explosion; another scenario is the use of a “dirty bomb” by terrorists. Nuclear weapons exposure is a significant health hazard; while the damage due to radiation from a dirty bomb could be significantly lower, there is still a threat of some type of radiation injury. In terms of internal radiation exposure to military personnel, the radioactive heavy metal depleted uranium (DU) is the most possible type of exposure. DU is used in military munitions and personnel can be wounded by DU shrapnel. A soldier wounded by internalized DU is considered to be carrying an internal emitter. While radioactive material that is inhaled or swallowed would be excreted from the system, embedded DU via a wound would not be rapidly excreted and presents a long-term hazard. These types of “battlefield” radiation exposures are unique to military personnel and are different than the high-dose fractionated therapeutic exposures that the average civilian might be exposed to during their lifetime.
High-dose radiation exposure can cause significant acute health effects including radiation syndrome and potentially death. Late health effects caused by radiation are also a significant health hazard. These late effects include cancer, leukemia, cytogenetic effects, and transgenerational effects to offspring. It is well known that radiation exposure can lead to cancer development and in particular to development of leukemia [1?4]. While there is significant human and animal data regarding the induction of high-dose radiation-induced cancers, less attention has been paid to whether the unique military radiation exposures like internalized DU or low-dose radiation potentially from dirty bombs can cause late health effects like cancer and genetic effects.

To better study the radiation late effects induced by the unique military radiation exposures a multi-parametric approach has been used in our laboratory at AFRRI. Implementation of cell- and animal-based multi-parametric assays can provide predictive information and be a guide as to whether human studies should be ursued. This strategic research approach involving the progression from cellular studies to animal model has been applied to radiation and heavy-metal studies in our laboratory (Figure 1).
Carcinogenic Hazard Evaluation
Transformation + Mutagenicity + Cytogenicity
Animal Carcinogenesis Model
Human Epidemiology
Figure 1: Short-term tests for carcinogenicity

2.0 LONG-TERM GOALS
The long-term research goals and benefits using the multi-parametirc approach are three-fold. First, the evaluation of potential carcinogenic exposures from radiation and/or DU in exposed individuals can be evaluated. Secondly, the development of nontoxic countermeasures to radiation-induced cancers can be undertaken. Thirdly, the potential discovery of biomarkers of exposure and disease development should be conducted simultaneously to the in vitro and in vivo cancer studies. The technical objectives of this approach include: 1) development of in vitro models to study radiation-induced late effects and radiobiology mech-anisms, and 2) development of in vivo models to study radiation-induced late effects and evaluate efficacy of pharmacological countermeasures.

3. 0 MULTI-PARAMETRIC RESEARCH APPROACH
The in vitro models that we have developed in our laboratory can be applied to everal research questions related to radiation late effects. These include: 1) neoplastic transformation models using human cells have been used to assess transformation; 2) the same transformation models have been used for rapid screening of pharmacological countermeasures; 3) a mutagenesis assay, the HPRT mutation assay (hypoxanthine guanine phosphoribosyl transferase), to measure mutagenesis has been established; 4) chromosomal aberration assays to measure chromosomal damage and genomic instability (potentially involved in carcinogenesis) have been
developed; and 5) clonal cell assays to measure radiobiology mechanisms, i.e., uranium “bystander” effects have been established.
Several of these assays can be used in combination to address the question as to whether a particular type of exposure is potentially carcinogenic. For example, our laboratory at AFRRI has used these in vitro assays to evaluate whether DU is carcinogenic. As shown in Figure 2, neoplastic transformation, mutagenicity, genotoxicity, and genomic instability were used to assay potential DU carcinogenicity. This slide contains data from our DU studies using human osteoblast cells (HOS) as our model system. Four endpoints were examined. These include mutagenicity, genotoxicity, neoplastic transformation, and genomic instability. A comparison was made to nickel and to external alpha particles. The DU exposure resulted in 17% of the cell nuclei being traversed by an alpha particle as measured by microdosimetry. In this multi-parametric in vitro approach, human cells are exposed to the test exposure/material and then plated for colony formation. An evaluation of colony morphology was used to define the state of transformation of the exposed cells. Further studies on the genotoxicity (measured as a sister chromatid exchange) and mutagenicity (measured as a mutation in the HPRT gene) were used in this multi-parameter approach.

Figure 2: In vitro assays to assess carcinogenic potential
These studies were the first to show that DU could transform human cells into the malignant phenotype and thus demonstrate the carcinogenic potential of DU. The additional assays including mutagenicity, geno-toxicity, and genomic instability, performed with this uranium compound further supported the finding the DU had carcinogenic potential even before animal or human studies were conducted. These in vitro models are an effective means to evaluate the carcinogenic potential of a particular type of exposure because they will enable the investigation to study a range of carcinogenic endpoints and provide rapid results.
This use of this multi-parametric approach is also effective for evaluating potential countermeasures to the late effects of radiation or internal emitters like DU. In particular the neoplastic transformation endpoint has been used to study potential radiation countermeasures. For example, as shown in Figure 3, our laboratory was able to preliminarily assess the efficacy of several pharmacological agents. In this assay we assessed the ability of these candidate drugs to inhibit malignant transformation in vitro. Data (transformation frequency) are shown in this figure for radiation (60Cobalt γ-rays) followed by a 15 day incubation with the candidate
agents. In this case we tested phenylactetate (PA), androstendiol (AED), and epigallocatechin gallate (EGCG), which were selected based on mechanistic considerations and low toxicity in vitro. Suppression of radiation-induced transformation was observed in cells treated with PA, AED, and EGCG under certain drug treatment conditions (drug exposure for 15 continuous days post-radiation).

Figure 3: Application of neoplastic transformation assay to screen potential chemoprevention agents
These data confirm that the in vitro transformation model can be used to screen for efficacy of potential radiation-chemopreventive agents and specifically indicate which of these agents should be next tested in the more laborious and expensive in vivo assays.
To evaluate potential late effects of radiation exposures to military personnel, in vivo models are also used. A multiparametric approach, similar to the in vitro approach, has been developed using several different animal models to study radiation carcinogenic effects. In contrast to in vitro models these in vivo models do not just focus on the development of late effects in the exposed cells or animals but are extended to an evaluation of how radiation exposure to one individual can affect another unexposed individual. Furthermore, these in vivo models enable a study of the most promising pharmacological countermeasures that were identified using the
in vitro methods. This multi-parametric in vivo approach involves the use of a leukemia model, a rapid trans-planted tumor model, and a transgenerational animal model to investigate offspring effects (Figure 4).

Development and application of in vivo models
1. Radiation- or depleted uranium-induced leukemia model in mice to study carcinogenesis and test late-effects countermeasures.
2. “Big blue” transgenic mouse model to study transgenerational radiation or DU effects on unexposed offspring.
3. Transplanted S-180 mouse tumor to rapidly screen pharmacological countermeasures rapidly (within 45 days).
Figure 4: Development and application of in vivo models First, a leukemia model is used to determine whether the type of exposure in questions can induce leukemia.
This model involves the injection of immature hematopoietic cells into irradiated or DU-exposed mice (Figure 5).
Figure 5: Radiation-induced leukemia DBA mice + (FDCPI cells) 60Cobalt gamma
The leukemia takes about 90?120 days to develop in irradiated animals. Control animals (un-irradiated) develop age-related leukemia only after approximately 300 days. This model is appropriate to evaluate radiation quality, radiation dose-rate, and radiation route-of-exposure effects, i.e., DU. For example our laboratory has used the model to examine and compare the effects of 60Cobalt γ-rays induced leukemia versus DU-induced leukemia (Figure 6) [5]. This leukemia model can also be further studied to evaluate the potential mechanisms involved in the leukemogenic process including epigenetic effects and the role of genomic instability. This model also has the potential to be used to assess the efficacy of late effects counter-measures since a comparison of the candidate drug to a vehicle can be conducted using this model.
Figure 6: DU carcinogenicity in vivo
An in vivo model to rapidly assess late effects radiation countermeasures is the transplanted tumor model. In this mode a lymphoma cell line is transplanted into the shoulder of a mouse and allowed to grow into a solid tumor. Within 21 days the tumor is palpable and can easily be measured. To test potential pharmacological countermeasures, the tumor is implanted, allowed to grow, and then the candidate drug (s) is administered. The effect on the size of the tumor is measured as a means to assess the efficacy of the drug. Results with PA and EGCG, previously identified as promising candidates with in vitro techniques, are shown in Figure 7. Thus a combination of the in vitro and in vivo methods has enabled us to identify the most promising coun-termeasures which can be further studied in more elaborate animal models.
Figure 7: Effect of EGCG and PA on tumor volumes of S-180 tumor-bearing mice
The third type of in vivo model used to assess radiation late effects involves a mouse model that enables us to determine whether radiation exposure to the parent can affect an unexposed offspring. It is well known that radiation exposure to a pregnant female can cause deleterious offspring effects during gestation [6]. Studies have demonstrated however, that parental preconceptional exposure (PPE) to certain qualities of radiation or heavy metal can induce cancer in unexposed offspring [7?9]. A transgenic mouse model is available that allows an investigation of whether parentral exposure causes offspring effects. This model, known as the
“Big Blue” is particularly effective in assessing whether a father’s exposure can affect unexposed offspring. It employs a transgenic mouse which has been engineered to carry the lacI regulatory gene in all tissues. Therefore, if a transgenic male is mated to a non-transgenic female mouse, approximately 50% of the offspring will carry the transgene, which they can only inherit from their transgenic father. The “Big Blue”
mutation model is appropriate to evaluate offspring responses in fathers exposed to the radiation or type of exposure of interest. Specifically, this model uses a mutation system [8] employing a λ shuttle vector carried by cells of a transgenic mouse (Stratagene Big Blue) that carries the target lacI gene. Big Blue male mice can be exposed to the exposure of interest (i.e., radiation, DU), mated with unexposed non-transgenic females and then the DNA can be recovered from the fathers and the tissues of the F1 offspring (Figure 8).
The recovered DNA is packaged and then assayed in vitro for mutations and detected in viral plaques by the blue color resulting from cellular metabolism. This model has been used in our laboratory to determine whether paternal exposure to DU can transmit genetic damage to the offspring. Bone marrow DNA from offspring identified as lacI carriers in treated and control groups was screened for mutations in the lacI transgene. Table 1 summarizes the bone marrow mutation frequencies found in the hemizygous F1 offspring of male parents exposed to DU, Ta, Ni, or 60Co radiation from the three experiments conducted. “Big Blue” Mutation and Offspring Assessment Assay

Figure 8: Model to assess transgenerational effects of radiation or heavy metals
Theses indicated that mutation frequencies from F1 offspring of DU-implanted fathers demonstrated a dose-dependent increase in comparison to control F1 offspring. Using this transgenic mouse model our laboratory was able to show that internalized DU exposure can cause adverse effects in unexposed offspring. This type of model is applicable to questions of parental exposure and potential deleterious offspring health effects. Table 1: Genotyping of offspring for transmission of the lacI gene Treatment


1
Based on expected 50% transmittal rate, the observed rates do not differ significantly (p > 0.05).
SUMMARY AND CONCLUSIONS
To better study the radiation late effects induced by the unique military radiation exposures, this multi-parametric approach has been used in our laboratory at AFRRI. Implementation of cell- and animal-based multi-parametric assays can provide predictive information and be a guide as to whether human studies should be pursued. This strategic research approach involving the progression from cellular studies to animal model has been applied to radiation and heavy metal studies in our laboratory and has provided a rapid and effective research approach to evaluating radiation risks for NATO and military personnel.
REFERENCES
1. Seed, T.M. Radiation Protectants: Current status and future prospects. Health Physics, 89(5):531?545, (2005).
2. Seed, T,M., Kumar, S., Whitnall, M., Venkataraman, S., Singh, V., Elliott, T., Landauer, M., Miller, A., Chang, C.-M., Inal, C., Deen, J., Genlhaus, M., Jackson III, W., Hilyard, E., Pendergrass, J., Toles, R.,
Villa, V., Miner, V., Stewart, M., Benjack, J., Danilenko, D., and Farrell, C., New strategies for the prevention of radiation Injury. J. Radiat. Res., 43: Suppl.:S239?S244, (2002).
3. Grdina, D.J., Murley, J.S., and Kataoka, Y. Radioprotectants: Current status and new directions. Oncology, 63 Suppl 2:2?10, (2002).
4. Dorr, R.T. Radioprotectants: pharmacology and clinical applications of amifostine. Semin. Radiat. Oncol., 8 (4 Suppl 1):10?13, (1998).
5. Miller, A.C. Leukemic transformation of hematopoietic cells in mice internally exposed to depleted uranium. Molecular Cellular Biochemistry 279(1-2):97?104, (2005).
6. Rugh R. The impact of radiation on the embryo and fetus. AJR, 89:181?190, 1963.
7. Gardner, M.J., Sneed, M.P., Hall, A.J., Powell, C.A., Downes, S., and Terrell, J.D. Results of case-control study of leukemia and lymphoma among young people near Sellafield nuclear plant in West
Cumbria. British Medical Journal. 300(6722):423?9, (1990).
8. Luke, G.A., Riches, A.C., Bryant, P.E., Genomic instability in hematopoietic cells of F1 generation mice of irradiated male parents. Mutagenesis 12(3):147?52, (1997).
9. Lord, B.I., Transgenerational susceptibility to leukemia induction resulting from preconception, paternal irradiation. Int. J. Radiat. Biol. 75(7):801?10, (1999).
10. Miller, A.C. Transgenerational Effects of Depleted Uranium, Health Physics, in press.
HFM Panel-099 RTG-033 Activity: Radiation Bioeffects and Countermeasures

RTG 033 Subgroup 1: Radiation Mechanisms and Late Effects

Chapter 3
Stimulation of the natural anti-tumour cells by single or fractionated irradiations of mice with X-rays

M.K. Janiak, E.M. Nowosielska, A. Cheda, J. Wrembel-Wargocka Military Institute of Hygiene & Epidemiology
4 Kozielska Str., Warsaw, Mazovia 01-163 Poland

1.0 INTRODUCTION
In the present day military scenarios involving radiation exposure the majority of the personnel is likely to absorb low to intermediate doses1 of predominantly low-LET2
ionising radiation [2]. Indeed, such doses will be incurred in areas of the enhanced radiation level due to the elevated natural background, contamination after explosions in nuclear installations or dispersal of radioactive material in the environment by other
means (radiation dispersal devices) and even after detonations of tactical nuclear bombs. Absorption of such doses will not evoke any of the acute post-irradiation effects but can potentially be associated with a long-term risk of subsequent cancers. Interestingly, however, accumulating evidence from the recent years indicates that absorption of doses of X- and γ-rays below 0.25 Gy may inhibit rather than exacerbate the development of various neoplasms [3?16].
One of the possible mechanisms of this effect is stimulation of anti-tumour immunity. Natural killer (NK) lymphocytes and activated cytotoxic macrophages are first-line effectors of the anti-neoplastic surveillance system. These cells non-specifically suppress the growth of tumour targets through secretion of a number of cytokines, such as perforins, granzymes, IL-2, IFN-γ, TNF-α, glutathione (NK lymphocytes) or IL-1, IL-12, TNF-α, GM-CSF, nitric oxide, and superoxide anions (activated macrophages). All these factors either directly induce apoptotic death of tumour cells or stimulate other cytolytic effector lymphocytes.
In view of the above, the aim of our study was to assess the effects of single and multiple low- and inter-mediate-level exposures to X-rays on the kinetics and mechanisms of non-specific cytotoxic reactions medi-ated by NK cells and/or macrophages and to correlate these effects with the anti-tumour activity of the irradiations.
We used male BALB/c mice aged 6?8 weeks. Peritoneal macrophages (Mφ) and NK cell-enriched spleeno-cytes (NK cells) obtained form the mice were irradiated in vitro with 0.1, 0.2, or 1.0 Gy X-rays or collected from the animals exposed to: a) single irradiation with 0.1, 0.2 or 1.0 Gy per mouse, or b) fractionated (5 days/week for 2 weeks) irradiation to obtain the absorbed doses of 0.01, 0.02 or 0.1 Gy per mouse per
1
According to the UNSCEAR 1986 Report [1], acute doses above 2 Gy, between 2 and 0.2 Gy, and below 0.2 Gy are regarded as high, intermediate, and low, respectively.
2
LET (Linear Energy Transfer) is a measure of the energy transferred to material as an ionizing particle travels through it. Typically this measure is used to quantify the effects of ionizing radiation on biological specimens.
fraction, so that total absorbed doses per mouse equalled to 0.1, 0.2 or 1.0 Gy, respectively. After the irradi-ations some mice were intravenously injected with syngeneic L1 sarcoma cells, sacrificed fourteen days later and tumour colonies were counted on the surface of the removed lungs. In the separated cell populations the following assays were carried out: a) cytotoxic activity and its suppression by specific inhibitors; b) blockade of the selected mechanisms of cytotoxicity; d) secretion of cytotoxic factors such as nitric oxide (NO), IL-1β, IL-2, IL-12, IFN-γ or TNF-α; e) apoptotic death.
2.0 RESULTS
2.1 Anti-tumour effects of low doses of X rays
The four separate experiments indicated that single whole body irradiation (WBI) of mice with 0.1 or 0.2 Gy led to the significant inhibition of the development of the pulmonary tumour colonies (expressed as percent of the control values measured in the sham-exposed animals). In contrast, no statistically significant reduction in the number of pulmonary tumour nodules could be detected when mice were pre-exposed to 1.0 Gy X-rays (Fig. 1A). The two consecutive experiments indicated that the fractionated WBI of mice with both 0.1 and 0.2 Gy X-rays resulted in the insignificant retardation of the development of pulmonary tumour colonies, whereas irradiation of mice with 1.0 Gy led to the slight increase in the number of the colonies (Fig. 1B).


Figure 1: Relative numbers (percentages of the control values indicated as solid line at 100%) of pulmonary L1 sarcoma cell colonies in mice exposed to single (A) or fractionated (B) 0.1, 0.2 or 1.0 Gy X-rays and two hours later i.v. injected with L1 sarcoma cells. Data are mean values ± SD. Results of four (A) or two (B) independent experiments are shown: each experimental group consisted of 12 mice. *?indicates statistically significant (p < 0.05) difference from the control (100%) value. Injection of the blockers of NK cells (anti-asialo GM1 antibody, anti-GM1 Ab) or macrophages (carrageenan,
CGN) almost totally eliminated the differences in the numbers of tumour colonies between the irradiated and control groups (Fig. 2). This effect was markedly more pronounced in the CGN- than in the anti-GM1 Ab-treated mice.

Figure 2: Relative numbers of pulmonary colonies after single WBI of mice and i.p. injection of anti-GM1Ab or CGN. C?sham-exposed, control mice; 0.1 Gy?mice exposed to a single WBI with 0.1 Gy X-rays; 0.2 Gy ? mice exposed to a single WBI with 0.2 Gy X-rays; 1.0 Gy?mice exposed to a single WBI with 1.0 Gy X-rays; PBS?mice i.p. injected with phosphate buffered saline; Ab?mice i.p. injected with anti-asialo GM1 antibody; CGN?mice i.p. injected with CGN. Presented are means ± SD from three independent experiments; each experimental group consisted of at least 12 mice. *?indicates statistically significant (p < 0.05) difference from the control/PBS (100%) value.
2.2 Mechanisms of the anti-neoplastic activity of NK lymphocytes
A single whole-body exposure of mice to any of the three doses of X-rays significantly stimulated the cytotoxic activity of NK cells, the effect being most pronounced on the second day after the irradiation.
Interestingly, the WBI of mice with 1.0 Gy (the dose that did not lead to inhibition of the growth of the pulmonary tumour nodules) appeared to be a more potent stimulator of the NK cell-mediated cytotoxicity than exposures to either 0.1 or 0.2 Gy X-rays (Fig. 3A). However, stimulatory effect of 1.0 Gy X-rays on the activity of NK cells could be partially explained by the possible elimination of radio-sensitive T and B cells
from the spleen leading to the relative increase in the percentage of the NK effectors in the cytotoxic assay (data not shown). In fact, as indicated by Lin et al. [17] and Harrington et al. [18] NK cells appear to exhibit the greatest radioresistance among the splenic lymphoid cells. Fractionated WBI of mice with either of the three applied doses of X-rays led to the significant enhancement of the cytotoxic function of NK cells to the comparable level in all the three groups (Fig. 3B).

Figure 3: Cytotoxic activity of NK cells tested on various days after the single (A) and fractionated (B) WBI of mice. C?sham-exposed, control mice; 0.1 Gy?mice exposed to WBI with 0.1 Gy X-rays;
0.2 Gy?mice exposed to WBI with 0.2 Gy X-rays; 1.0 Gy?mice exposed to WBI with 1.0 Gy X-rays. Mean values ± SD obtained from three independent experiments are presented; each experimental group consisted of five mice. When mice were injected with the anti-GM1 Ab, the activity of these cells, as tested 2 days later, was totally abrogated. This inhibition could not be reversed by WBI with 0.1 or 0.2 Gy X rays (Fig. 4).


Figure 4: Cytotoxic activity of splenic NK cells (at 100:1 E:T ratio) on the second day after irradiation of mice with 0.1, 0.2 or 1.0 Gy X-rays. C?sham-exposed, control mice; 0.1 Gy?mice exposed to a single WBI with 0.1 Gy X-rays; 0.2 Gy?mice exposed to a single WBI with 0.2 Gy X-rays; 1.0 Gy? mice exposed to a single WBI with 1.0 Gy X-rays; NK?mice i.p. injected with PBS;
NK+Ab?mice injected with anti-asialo GM1 Ab. Presented are means ± SD from three independent experiments; each experimental group consisted of at least three mice. ^?indicates statistically
significant (p<0.05) difference from the control value; *?indicates statistically significant (p<0.05) difference within sham-irradiated and irradiated groups between mice injected with PBS and mice
injected with anti-asialo GM1 Ab.
The elevated cytolytic activity of NK lymphocytes after irradiation of mice with all the three doses of single or fractionated X-rays was, for the most part, mediated by the perforin and the Fas receptor ligand (FasL) pathways (Fig. 5). This was corroborated by the finding that NK cells obtained from mice 2 days after the
single WBI with 0.1 and 0.2 Gy X-rays (i.e. at the time when the cytotoxic function of these cells was maximally stimulated) demonstrated the significantly increased surface expression of FasL as compared to
the cells collected form the sham-exposed animals. No such effect was detected after the irradiation of mice with 1.0 Gy X-rays (Fig. 6).

Figure 5: Inhibition of the NK-type cytotoxic activity of splenocytes by CMA and the anti-FasL Ab on the 2nd day after the single (A) and on the 3rd day after the fractionated (B) WBI of mice. C?sham-exposed, control mice; 0.1 Gy?mice exposed to WBI with 0.1 Gy X-rays; 0.2 Gy?mice exposed to WBI with 0.2 Gy X-rays; 1.0 Gy?mice exposed to WBI with 1.0 Gy X-rays; NK?NK cells incubated without blockers; NK+CMA?NK cells incubated with CMA; NK+ anti-FasL?NK cells incubated with anti-FasL antibody; NK+CMA+anti-FasL? NK cells incubated with CMA and anti-FasL antibody. Mean values ± SD obtained from three independent experiments are presented; each experimental group consisted of five mice. ^?indicates statistically significant (p<0.05) difference between NK cells collected from irradiated mice and the respective NK cells obtained from non-irradiated mice. *?indicates statistically significant (p<0.05) difference within sham-
irradiated or irradiated groups between NK cells incubated with CMA and/or anti-FasL antibody and NK cells incubated without blockers.

Figure 6: Relative (percentage of the control value indicated as solid line at 100%) surface expression of FasL on NK cells two days after a single WBI of mice with 0.1, 0.2 or 1.0 Gy X-rays.
C?sham-exposed, control mice; 0.1 Gy?mice exposed to a single WBI with 0.1 Gy X-rays; 0.2 Gy?


mice exposed to a single WBI with 0.2 Gy X-rays; 1.0 Gy ?mice exposed to a single WBI with 1.0 Gy X-rays. Mean values ± SD obtained from three independent experiments are presented; each experimental group consisted of three mice. *?indicates statistically significant (p<0.05) difference from the control (100%) value.
However, both concanamicin A (CMA), a blocker of perforin, as well as the anti-FasL antibody were unable to totally suppress the cytolytic function the NK cells. These results suggested that the residual cytotoxic activity of the effector cell populations might be due to synthesis and/or secretion of additional cytotoxic and/or cytostatic factors likely to be involved in elimination of neoplastic cells. Indeed, we demonstrated that both single and fractionated WBI of mice with all the three doses of X-rays significantly stimulated synthesis of IL-2 and IFN-γ in the splenocytes and the NK cells, respectively (Figs. 7 and 8). IL-2 is a prominent activator of cytotoxic T and NK lymphocytes [19,20] whereas IFN-γ, although usually not directly cytocidal for tumour cells, stimulates cytolytic functions of macrophages and, together with the macrophage-derived TNF-α and IL-1β, can exert a strong anti-neoplastic effect [21,22].

Figure 7: Production of IL-2 by splenocytes after single (A) or fractionated (B) WBI of mice and incubation with PHA. C?sham-exposed, control mice; 0.1 Gy?mice xposed to WBI with 0.1 Gy X-rays; 0.2 Gy?mice exposed to WBI with 0.2 Gy X-rays; 1.0 Gy?mice exposed to WBI with 1.0 Gy X-rays. Mean values ± SD obtained from three independent experiments are presented; each experimental group consisted of three mice.

Figure 8: Production of IFN-γ by NK cells after single (A) or fractionated (B) WBI of mice and incubation with YAC-1 cells. C?sham-exposed, control mice; 0.1 Gy?mice exposed to WBI with 0.1 Gy X-rays; 0.2 Gy?mice exposed to WBI with 0.2 Gy X-rays;1.0 Gy?mice exposed to WBI with 1.0 Gy X-rays. Mean values ± SD obtained from three independent experiments are presented; each experimental group consisted of three mice. Irradiation of mice with 0.1 or 0.2 Gy of X-rays did not affect the rate of apoptosis in the examined NK cell-suspensions, whereas similar irradiations with 1.0 Gy enhanced the number of apoptotic NK lymphocytes (Fig. 9).

Figure 9: Apoptosis of NK cells after WBI of mice. C?sham-exposed, control mice; 0.1 Gy?mice exposed to WBI with 0.1 Gy X-rays; 0.2 Gy?mice exposed to WBI with 0.2 Gy X-rays ;1.0 Gy?mice exposed to WBI with 1.0 Gy X-rays. Mean values ± SD obtained from three independent experiments are presented; each experimental group consisted of three mice.
2.3 Mechanisms of the anti-neoplastic activity of peritoneal macrophages The three separate experiments indicated that a single WBI of mice with either 0.1 or 0.2 Gy X-rays led to the significant elevation of the cytotoxic activity of the IFN-γ- and LPS-boosted Mφ against the L1 tumour cells, the effect being most pronounced between the third and fifth days post-exposure to X-rays. In contrast,
a single WBI of mice with 1.0 Gy did not affect the cytotoxic function of Mφ (Fig. 10A).
Fractionated WBI of mice with either of the three applied doses of X-rays resulted in the significant enhance-ment of the cytotoxic activity of Mφ to the same level in all the three groups, the effect being most pro-nounced between the second and fifth days post-irradiation and then declined (Fig. 10B).

Figure 10: Cytotoxic activity of the IFN-γ- and LPS-treated Mφ after single (A) or fractionated (B) WBI of mice and incubation with L1 cells. C?sham-exposed, control mice; 0.1 Gy?mice exposed to WBI with 0.1 Gy X-rays; 0.2 Gy?mice exposed to WBI with 0.2 Gy X-rays; 1.0 Gy?mice exposed to WBI with 1.0 Gy X-rays. Mean values ± SD obtained from three independent experiments are presented; each experimental group consisted of five mice.
The stimulated Mφ-mediated cytolysis of susceptible tumour targets was for the most part associated with the elevated production of nitric oxide (NO) that expressed similar kinetics as the cytotoxic activity of these cells (Fig. 11).

Figure 11: Production of NO by the IFN-γ- and LPS-treated Mφ after single (A) or fractionated (B) WBI of mice. C?sham-exposed, control mice; 0.1 Gy?mice exposed to WBI with 0.1 Gy X-rays; 0.2 Gy?mice exposed to WBI with 0.2 Gy X-rays; 1.0 Gy?mice exposed to WBI with 1.0 Gy X-rays. Mean values ± SD obtained from three independent experiments are presented; each experimental group consisted of five mice.
Mφ collected from mice pre-injected with CGN were significantly less cytotoxic against the L1 cells than macrophages obtained from the CGN-untreated animals. Even more pronounced reduction of the cytotoxic
function of Mφ was detected in the cells obtained from the CGN-untreated mice and incubated in vitro in the presence of aminoguanidine (AG) (Fig. 12A). Also, production of NO by Mφ obtained from both the sham-
and X-ray-exposed mice pre-treated with CGN was almost totally suppressed in all the groups. As expected, addition of AG to the incubation medium of the collected Mφ led to the significant inhibition of the production of NO whose level was even lower than in cells collected from the CGN-treated mice (Fig. 12B).

Figure 12: CGN- and AG-induced suppression of cytotoxic activity (A) and production of NO (B) by the IFN-γ- and LPS-treated (S) Mφ on the third day after single WBI of mice. C?sham-exposed, control mice; 0.1 Gy?mice exposed to WBI with 0.1 Gy X-rays; 0.2 Gy?mice exposed to WBI with 0.2 Gy X-rays ;1.0 Gy?mice exposed to WBI with 1.0 Gy X-rays. M + S?Mφ obtained from the CGN-untreated mice; M + S + CGN?Mφ obtained from mice pretreated with CGN; M + S + AG?Mφ obtained from the CGN-untreated mice and incubated in vitro in the presence of AG. M + S + CGN + AG?Mφ obtained from mice pretreated with CGN and incubated in vitro in the presence of AG.
Mean values ± SD obtained from two independent experiments are presented; each experimental group consisted of five mice ^?indicates statistically significant (p<0.05) difference between Mφ collected from irradiated mice and the respective Mφ obtained from non-irradiated mice. *?indicates statistically significant (p<0.05) difference from the results obtained in group M + S.
However, neither i.p. injection of mice with CGN, a lysosome-disrupting and phagocyte-damaging com-pound, nor addition to the culture medium of AG, a classical inhibitor of the inducible NO synthase, totally abrogated the cytotoxic activity of Mφ even when both blockers were used concurrently. These results sug-gested that the residual cytotoxic activity of the effector cell populations might be due to synthesis and/or secretion of additional cytotoxic and/or cytostatic factors that are likely to be involved in elimination of neoplastic cells. Indeed, our investigations demonstrated that both single and fractionated irradiations of mice with all the three applied doses of X-rays significantly stimulated Mφ to produce a number of cytokines with potential anti-neoplastic properties. These include IL-1β, IL-12, TNF-α (Figs. 13?15).

Figure 13: Production of IL-1β by the IFN-γ- and LPS-treated Mφ after single (A) or fractionated (B)
WBI of mice and incubation with L1 cells. C?sham-exposed, control mice; 0.1 Gy?mice exposed to WBI with 0.1 Gy X-rays; 0.2 Gy?mice exposed to WBI with 0.2 Gy X-rays; 1.0 Gy?mice exposed
to WBI with 1.0 Gy X-rays. Mean values ± SD obtained from three independent experiments are presented; each experimental group consisted of five mice.

Figure 14: Production of IL-12 by the IFN-γ- and LPS-treated Mφ after single (A) or fractionated (B)
WBI of mice and incubation with L1 cells. C?sham-exposed, control mice; 0.1 Gy?mice exposed to WBI with 0.1 Gy X-rays; 0.2 Gy?mice exposed to WBI with 0.2 Gy X-rays; 1.0 Gy?mice exposed
to WBI with 1.0 Gy X-rays. Mean values ± SD obtained from three independent experiments are presented; each experimental group consisted of five mice.

Figure 15: Production of TNF-α by the IFN-γ- and LPS-treated Mφ after single (A) or fractionated (B)
WBI of mice and incubation with L1 cells. C?sham-exposed, control mice; 0.1 Gy?mice exposed to WBI with 0.1 Gy X-rays; 0.2 Gy?mice exposed to WBI with 0.2 Gy X-rays; 1.0 Gy?mice exposed
to WBI with 1.0 Gy X-rays. Mean values ± SD obtained from three independent experiments are presented; each experimental group consisted of five mice.
2.4 NK cell- and macrophage-mediated activity after single in vitro irradiations Unlike the low-level irradiations of mice, single exposures of the isolated NK cells and Mφ to either of the
applied single doses of X-rays did not boost the cytotoxic activities of these cells (Table 1).

Table 1: Activity of NK cells and IFN-γ- and LPS-treated Mφ after in vitro irradiation. C?sham-exposed, control cells; 0.1 Gy?cells exposed to 0.1 Gy X-rays; 0.2 Gy?cells exposed to 0.2 Gy X-
rays; 1.0 Gy? cells exposed to 1.0 Gy X-rays. Mean values ± SD obtained from two independent experiments are presented; each experimental group consisted of five mice.
Irradiation of the isolated NK cells with 0.1 or 0.2 Gy X-rays did not affect the rate of apoptosis in the examined cell suspensions, whereas irradiation with 1.0 Gy enhanced the number of apoptotic NK cells (Fig. 16).

Figure 16: Apoptosis of NK cells after in vitro irradiation. C?sham-exposed, control cells; 0.1 Gy? cells exposed to 0.1 Gy X-rays; 0.2 Gy?cells exposed to 0.2 Gy X-rays; 1.0 Gy?cells exposed to
1.0 Gy X-rays. Mean values ± SD obtained from two independent experiments are presented; each experimental group consisted of five mice.
The in vitro irradiations of Mφ with 0.1, 0.2, or 1.0 Gy X-rays did not affect the rate of apoptosis (Fig. 17).

Figure 17: Apoptosis of IFN-γ- and LPS-treated Mφ after in vitro irradiation C?sham-exposed, control cells; 0.1 Gy? cells exposed to 0.1 Gy X-rays; 0.2 Gy?cells exposed to 0.2 Gy X-rays; 1.0
Gy?cells exposed to 1.0 Gy X-rays. Mean values ± SD obtained from two independent experiments are presented; each experimental group consisted of five mice.

3. 0 CONCLUSIONS
The obtained data indicate that:
1. Both single and fractionated whole-body exposures of mice to low (0.1 and 0.2 Gy) but not intermediate (1.0 Gy) doses of X-rays inhibit (single irradiation) or tend to inhibit (fractionated irradiation) the
development of the induced pulmonary tumour colonies;
2. The suppression may result from stimulation of the natural defence reactions mediated by NK lymphocytes and/or cytotoxic macrophages;
3. Boosting of the cytolytic functions of the above anti-neoplastic effectors by low-level exposures to X-rays requires the presence of other cells and/or environmental factors available in the in vivo but not the
in vitro conditions.
4.0 REFERENCES
[1] UNSCEAR (1986) United Nations scientific committee on the effects of atomic radiation, 1986 Report to the general assembly, with annexes. Genetic and somatic effects of ionizing radiation United
Nations Publ. E.86.IX, UN, New York, p 170.
[2] Potential radiation exposure in military operations. Protecting the soldier before, during and after (1999), Thaul S, O’Maonaigh H (Eds) Institute of Medicine, National Academy Press Washington D.C.
[3] Hashimoto S, Shirato H, Hosokawa M, Nishioka T, Kuramitsu Y, Matushita K, Kobayashi M, Miyasaka K (1999) The suppression of metastases and the change in host immune response after low-
dose total-body irradiation in tumor-bearing rats. Radiat Res 151:717?724.
[4] Ju GZ, Liu SZ, Li XY, Liu WH, Fu HQ (1995) Effect of high versus low dose radiation on the immune system. In: Hagen U, Harder D, Jung H, Streffer C (eds) Radiation research 1895?1995. Proceedings
on tenth international congress of radiation research, Wurzburg, Germany, 27 August September 1995. IARR, Wurzburg, pp 709 714.
[5] Kojima S, Ishida H, Takahashi M, Yamaoka K (2002) Elevation of glutathione induced by low-dose gamma rays and its involvement in increased natural killer activity. Radiat Res 157:275?280.
[6] Liu SZ, Xiao PX, Ma SY, Xu GZ, Tian CH, Yu HY, Zhang LM (1982) A study of the immune status of inhabitants in an area of high natural radioactivity in Guangdong. Chin J Radiol Med Prot 2:64?68.
[7] Liu SZ, Xu GZ, Li XY, Xia FQ, Yu HY, Qi J, Wang FL, Wang SK (1985) A restudy of immune functions of the inhabitants in a high natural radioactivity area in Guangdong. Chin J Radiol Med Prot 5:124?127.
[8] Liu SZ (1989) Radiation hormesis. A new concept in radiological science. Chin Med J 102:750?755.
[9] Liu SZ, Su X, Zhang YC, Zhao Y (1994) Signal transduction in lymphocytes after low dose radiation. Chin Med J 107:431?436.
[10] Liu SZ, Zhang YC, Mu Y, Su X, Liu JX (1996) Thymocyte apoptosis in response to low-dose irradiation. Mutat Res 358:185?191.
[11] Liu SZ (2004) Cancer control related to stimulation of immunity by low dose radiation. In: Proceedings of 14th pacific basin nuclear conference, Honolulu, HI, 21?25 March 2004. American
Nuclear Society, La Grange Park, pp 368?372.
[12] Luckey TD (1999) Nurture with ionising radiation: a provocative hypothesis. Nutr Cancer 34:1?11
[13] Pandey R, Shankar BS, Sharma D, Sainis KB (2005) Low dose radiation induced immunomodulation: effect on macrophages and CD8+ T cells. Int J Radiat Biol 81:801?812.
[14] Safwat A (2000) The immunology of low-dose total-body irradiation: more questions than answers. Radiat Res 153:599?604.
[15] Safwat A (2000) The role of low-dose total body irradiation in treatment of non-Hodgkin’s lymphoma: a new look at an old method. Radiother Oncol 56:1?6.
[16] Safwat A, Bayoumy Y, El-Sharkawy N, Shaaban K, Mansour O, Kamel A (2003) The potential palliative role and possible immune modulatory effects of low-dose total body irradiation in relapsed or chemo-resistant non-Hodgkin’s lymphoma. Radiother Oncol 69:33?36.
[17] Lin IH, Hau DM, Chen WC, Chen KT (1996) Effects of low dose gamma-ray irradiation on peripheral leukocyte counts and spleen of mice. Chin Med J 109:210?214.
[18] Harrington NP, Chambers KA, Ross WM, Filion LG (1997) Radiation damage and immune suppression in splenic mononuclear cell populations. Clin Exp Immunol 107:417?424.
[19] DeBlaker-Hohe DF, Yamauchi A, Yu CR, Horvath-Arcidiacono JA, Bloom ET (1995) IL-12 synergises with IL-2 to induce lymphokine-activated cytotoxicity and perforin and granzyme gene expression in fresh human NK cells. Cell Immunol 165:33?43.
[20] Miller GM, Kim DW, Andres ML, Green LM, Gridley DS (2003) Changes in the activation and reconstitution of lymphocytes resulting from total-body irradiation correlate with slowed tumor
growth. Oncology 65:229?241.
[21] Al-Sarireh B, Eremin O (2000) Tumour-associated macrophages (TAMS): disordered function, immune suppression and progressive tumour growth. J R Coll Surg Edinb 45:1?16.
[22] Belardelli F, Ferrantini M (2002) Cytokines as a link between innate and adaptive antitumour immunity. Trends Immunol 23:201?208.


02. 2012年7月17日 01:35:58 : pi0fYycp4s
Chapter 4
German Contributions Using Gene Expression Studies to Detect Radiation Targets and to Discriminate Radiation Associated Tumors from Other Ethiologies

Matthias Port
1
, Christian G. Ruf
2
, Armin Riecke
2
and Michael Abend2


1
Department of Hematology, Hemostaseology, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
2
Bundeswehr Institute of Radiobiology, German Armed Forces, Munich, Germany

ABSTRACT
Within this chapter we describe nowadays used modern approaches for detection of radiation induced gene targets employing whole genome microarrays for screening purposes of candidate genes and their quanti-
tative analysis using RTQ-PCR (two stage study design). Besides this methodological focus (also including results on normalization procedures) we describe gene expression examinations which allow to discriminate
radiation associated thyroid cancer tumors from those developed due to other etiologies which might be of importance in the context of compensation requests occurring after e.g. accidental or occupational radiation
exposures.
1.0 APPROACHES FOR DETECTION OF RADIATION INDUCED GENE TARGETS
1.1 Combining whole genome microarray with RTQ-PCR Recently Cam and coworker hybridized one cDNA probe on three different microarray platforms in order to
compare the overall agreement among these platforms. A total of 4 from 185 genes tested were similarly detected with all three microarrays [Tan et al. 2003]. This was another experiment raising doubts about gene
expression analysis using microarrays and about the reproducibility of this method. In 2005 the so called “MicroArray Quality Control” (MAQC) project was initiated by the U.S. Food and Drug Admininistration
[Casciano and Woodcock 2006]. A consortium of 137 researchers from 51 different organizations examined reproducibility across laboratories by putting 20 microarray products and three alternative technologies
through more than 1,300 tests [Couzin 2006].
It is widely accepted using real-time quantitative PCR (RTQ-PCR) as a reference method for quantification of RNA-copy numbers [Canales et al. 2006, Mackay et al. 2002, Wang et al. 2006]. That holds true
especially due to its advantages in detection sensitivity, sequence specificity, large dynamic range as well as its high precision and reproducible quantitation compared to other techniques [Wong and Medrano 2005, Wilhelm and Pingoud 2003].
In general small amounts of RNA are amplified before doing the hybridization on the microarray. In previous experiments we showed a high variability inherent to this step. As an alternative we pooled RNA
from several experiments. This allowed reducing the RNA needed for a complete gene expression experi-ment including RTQ-PCR validation down to less than 5 μg per individual. Furthermore we demonstrate that
when methods are well established in a laboratory one array and RTQ-PCR measurements performed only in duplicates reaching effectiveness and reliability comparable to the results of the MAQC consortium (sub-
mitted for publication).
1.2 Normalization of gene expression using RTQ-PCR
A large number of housekeeping genes for normalization purposes of gene expression data exist. Companies even offer platforms (96-well format) in order to find out about the appropriate gene or combination of genes.

Figure 1: Comparison of absolute gene expression of 3 house-keeping genes in 3 tissue types of 9 human individuals. Bars represent the mean calculated from 3 individuals. Error bars show the
SEM, n=3 (modified graph published by Port et al. 2007a).
Depending on the experimental design (kind and intensity of exposure, endpoint, biological sample and time) the desired gene might differ. Among many other genes 18S ribosomal RNA (18S rRNA) is widely used for
normalization purposes.
We in particular examined the usefulness of known housekeeping genes (Figure 1) for normalization purposes after ionising radiation in different models and points in time after radiation (Figure 2).
In summary, 18S rRNA proved its superiority over other widely used genes for normalization purposes after radiation exposure in a large variety of biological models [Port et al. 2007a].
1.3 CLARCC array, a quantitative gene expression platform for known radiation induced signal transduction pathways Recently the dependency in the mode of cell death on three parameters has been demonstrated. These
parameters include the kind and intensity of the stressor as well as the irradiated model [Abend et al. 1995, 1996, 2000]. Depending on these parameters a shift in the mode of cell death happens [Abend 2003].
Not only apoptosis, but also certain kind of necrosis and even the process of micronucleation or mitotic catastrophe seem to be regulated by the cell on the gene expression level (Stassen et al. 2003, Seidl et al. 2007).

Figure 2: Comparing differential gene expression (relative to control) in two irradiated in vitro cell lines up to 48 h after radiation exposure and in normal tissues of 3 tissue types of 9 human individuals,
human testis tumors and different dog tissues [modified graphs published by Port et al. 2007a].
A RTQ-PCR based low density array was designed (CLARCC array, acronym for cell cycle, lipid metabolism, apoptosis, repair and cytokinesis & chromosome segregation, Figure 3). With altogether 380
genes measured simultaneously it covers the nowadays most important known genes coding for the bio-logical processes including different modes of cell death (lipid metabolism, apoptosis and cytokinesis &
chromosome segregation), proliferation and repair (e.g. dsb). Moreover, since the platform chosen is a RTQ-PCR technique it represents the gold standard for gene expression measurements leading to quantitative and
not only semiquantitative results of several hundred genes at the same time.
Recently, the CLARCC array was used to examine a gene expression “fingerprint” on a human gastric tumor cell line (HSC) exposed to an alpha-emitting radionuclide (Seidl et al. 2007). Actually it is used on HL-60
cells (model for apoptosis) after gamma ray exposure.

Figure 3: Description of gene categories associated to biological processes covering cell cycle, lipid metabolism, apoptosis, repair and cytokinesis & chromosome segregation (CLARCC array).
2.0 DISCRIMINATING RADIATION ASSOCIATED TUMOURS FROM OTHER
ETIOLOGIES
In general, the cause of a developing tumor and in particular the causal link to ionizing radiation cannot be made. However, due to possible compensation requests of exposed individuals it is desirable to be able to do that.
For this reason, pooled RNA from 10 tumour tissues (papillary thyroid cancer, PTC) of Belarusian patients (total number of patients was 11) subjected to radiation after the Chernobyl nuclear accident and pooled
RNA from 10 individuals of a control group consisting of 41 Caucasian patients originating from south-eastern Germany and suffering from thyroidal carcinoma of similar histology but lacking a radiation
exposure history were hybridised on a whole genome microarray for screening of potentially upregulated or downregulated genes. Results of microarrays are semiquantitative and must be validated with another
method, preferably quantitative real time polymerase chain reaction (RTQ-PCR), the well accepted, convenient and economical method to confirm array data for gene expression measurements. Nearly 100 of
the most promising genes screened with the microarray were examined quantitatively on each of the biopsies with a recently developed RTQ-PCR-based technology called low-density array (LDA) in order to identify
the most promising genes for distinguishing between sporadic and radiation-induced PTC.

Table 1: Characterization of the seven most promising gene targets for distinguishing between post-Chernobyl PTC and a control group lacking an additional radiation exposure history.
Information were taken from NCBI Entrez Gene database (updated Mar 2006). Only literature associating the genes with tumour development was cited. The abbreviation “n.e.” means “no
entry” in the database. Data were taken from published work [Port et al. 2007b].
In summary, our microarray data on post-Chernobyl PTC reflect a positive stimulus for tumor growth (overrepresentation of upregulated genes coding for G-proteins and growth factor) and increased
aggressiveness caused by overexpressed oxidoreductases, while the immune defense (overrepresentation of downregulated genes coding for immunglobulin) appeared weakened. For the first time, a complete
differentiation of post-Chernobyl PTC from controls either characterized by comparable aggressiveness (PTC of older patients, median age: 60 years) or comparable age (n =4) was accomplished by a selection of
seven gene targets (Table 1). Further examinations on larger groups are needed in order to determine whether these findings are applicable as a diagnostic tool for identifying radiation-induced PTC.
3. REFERENCES
[1] Abend, M., Rhein, A., Gilbertz, K.P., Blakely, W.F., van Beuningen, D. Correlation of micronucleus and apoptosis assays with reproductive cell death. Int. J. Radiat. Biol. 67(3):315?26, (1995).
[2] Abend, M., Rhein, A., Gilbertz, K.P., van Beuningen, D. Evaluation of a modified micronucleus assay.
Int. J. Radiat. Biol. 69(6):717?27, (1996).
[3] Abend, M., Kehe, K., Kehe, K., Riedel, M., van Beuningen, D. Correlation of micronucleus and apop-tosis assays with reproductive cell death can be improved by considering other modes of death. Int. J.
Radiat. Biol. 76(2):249?59, (2000).
[4] Abend, M. Reasons to reconsider the significance of apoptosis for cancer therapy. Int. J. Radiat. Biol. 79:927?941,(2003).
[5] Casciano, D.A., Woodcock, J. Empowering microarrays in the regulatory setting. Nat. Biotechnol. 24(9):1103, (2006).
[6] Couzin, J. Genomics. Microarray data reproduced, but some concerns remain. Science. 313(5793):1559, (2006).
[7] Canales, R.D., Luo, Y., Willey, J.C., Austermiller, B., Barbacioru, C.C., Boysen, C., Hunkapiller, K., Jensen, R.V., Knight, C.R., Lee, K.Y. et al.: Evaluation of DNA microarray results with quantitative
gene expression platforms. Nat. Biotechnol. 24:1115?1122, (2006).
[8] Mackay, I.M., Arden, K.E., Nitsche, A. Real-time PCR in virology. Nucleic Acids Res. 30:1292?1305, (2002).
[9] Port, M., Schmelz, H.U., Stassen, T., Mueller, K., Stockinger, M., Obermair, R., Abend, M.. Correcting false gene expression measurements from degraded RNA using RTQ-PCR. Diagn. Mol.
Pathol. 16(1):38?49, (2007a).
[10] Port, M., Boltze, C., Wang, Y., Roper, B., Meineke, V., Abend, M. A radiation-induced gene signature distinguishes post-Chernobyl from sporadic papillary thyroid cancers. Radiat Res. 168(6): 639?49,
(2007b).
[11] Seidl, C., Port, M., Gilbertz, K.P., Morgenstern, A., Bruchertseifer, F., Schwaiger, M., Roper, B., Senekowitsch-Schmidtke, R., Abend, M. 213Bi-induced death of HSC45-M2 gastric cancer cells is
characterized by G2 arrest and up-regulation of genes known to prevent apoptosis but induce necrosis and mitotic catastrophe. Mol. Cancer. Ther. 6(8):2346?59, (2007).
[12] Stassen, T., Port, M., Nuyken, I., Abend, M. Radiation-induced gene expression in MCF-7 cells. Int. J. Radiat. Biol. 79(5):319?31, (2003).
[13] Tan, P.K., Downey, T.J., Spitznagel, E.L. Jr, Xu, P., Fu, D., Dimitrov, D.S., Lempicki, R.A., Raaka, B.M., Cam, M.C. Evaluation of gene expression measurements from commercial microarray
platforms. Nucleic Acids Res. 31(19):5676?84, (2003).
[14] Wang, Y., Barbacioru, C., Hyland, F., Xiao, W., Hunkapiller, K.L., Blake, J., Chan, F., Gonzalez, C., Zhang, L., Samaha, R.R. Large scale real-time PCR validation on gene expression measurements from
two commercial long-oligonucleotide microarrays. BMC Genomics 7:59, (2006).
[15] Wong, M.L., Medrano, J.F. Real-time PCR for mRNA quantitation. Biotechniques. 39(1):75?85, (2005).
[16] Wilhelm, J., Pingoud, A. Real-time polymerase chain reaction. Chembiochem. 4(11):1120?8, (2003).

Chapter 5
Molecular Biomarkers of Acute Radiation Syndrome and Radiation Injury William F. Blakely,Gregory L. King,Matthias Port, and Natalia I. Ossetrova1

Armed Forces Radiobiology Research Institute Uniformed Services University
8901 Wisconsin Avenue Bethesda, MD 20889-5603 USA
and Department of Hematology, Hemostaseology, Oncology and Stem Cell Transplantation Hannover Medical School Hannover, Germany


ABSTRACT
The acute radiation syndrome or sickness (ARS) represents signs and symptoms associated with various organ and tissues systems response after exposure to radiation. Here we review the current status of
knowledge on the use of molecular and other metabolic biomarkers, based on specific organs and tissue systems, as candidate bioindicators for radiation injury severity. An assessment of the confounders and
research gaps for diagnostic use of molecular biomarkers, in complementation with conventional diagnostic methodologies, in the development of medical treatment decisions is discussed.
1.0 INTRODUCTION
Acute radiation syndrome or sickness (ARS) constitutes a constellation of signs and symptoms from several organ subsystems (i.e., cerebrovascular, hematopoietic system, gastrointestinal or GI, cutaneous, respiratory,
etc.), each exhibiting distinct time- and dose-dependent responses from the radiation injury [Waselenko et al. 2004; Gorin et al. 2006]. Effective medical management of a suspected radiation-overexposure patient
necessitates recording dynamic medical clinical data, measuring appropriate radiation bioassays, and estimating dose from dosimeters and radioactivity assessments in order to provide diagnostic information to
the treating physician and dose assessment for personnel radiation-protection records. The accepted generic multiparameter approach includes measuring radioactivity and monitoring the exposed individual; observing
and recording prodromal signs/symptoms including erythema; obtaining complete blood counts (CBC) with white blood cell differential; sampling blood for the chromosome-aberration cytogenetic bioassay using the
―gold standard? dicentric assay for dose assessment and dose in-homogeneity (whole- vs. partial-body exposure); bioassay sampling, if appropriate, to determine radioactivity contamination; and using other
available dosimetry [Blakely et al. 2005] and clinical approaches. Professor Fliedner and colleagues (University of Ulm, Germany) have developed an ARS severity scoring system based on clinical signs and
symptoms for the major subsyndromes (i.e., neurovascular, gastrointestinal, hemopoietic, cutaneous, etc.).
Called ― Medical Treatment Protocols (METREPOL) it presents a grading score to quantify the severity level for the various subsyndromes of ARS (Fliedner et al. 2001) and forms the basis of a medical treatment
decision guidance. AFRRI recently updated it Biological Dosimetry worksheet, available on AFRRI’s website (www.afrri.usuhs.mil), to include a modified version of METREPOL.
Biochemical or molecular biomarkers represent an additional complementary diagnostic approach that has the potential to provide information on radiation injury and hence formulate medical treatment decisions. For
example, blood plasma or serum biochemical markers of radiation exposure have been advocated for use in early triage and injury assessment of radiation casualties [Bertho et al. 2001; Blakely et al. 2003a, 2003b;
Roy et al. 2005; Marchetti et al. 2006; Ossetrova et al. 2007, 2009, 2010; Blakely et al. 2007, 2010; Okunieff et al. 2008; Guipaud and Bendritter et al. 2009; Ossetrova and Blakely 2010] (Table 1). Biomarkers can fall
into two classes, early expressed biomarkers of radiation injury as well as organ-specific injury biomarkers that are exhibited at various times after radiation exposure in a time- (Figure 1) and dose-dependent (Figure
2) fashion based on organ- and tissue-specific cell-renewal transit times [Hall and Giaccia, 2006], which have been validated in several radiation models, see Table 2.

The blood plasma or serum biomarker approach has several advantages. Early biomarkers may contribute along with other biodosimetric indices, clinical signs and symptoms, and evidence of physical dose to initiate use of
non-toxic medical countermeasures that demonstrate greater efficacy when started 24 h after radiation exposure [MacVittie et al. 2005]. Organs and tissues leak tissue and organ specific bio-indicators into blood when
responding to radiation damage, so their measurements in blood can provide useful diagnostic information about the severity of specific organ and tissue system to radiation injury. In this report, we review the current
status of use of molecular and other biomarkers of tissues and organs systems associated with ARS and radiation injury. We also identify some potential major confounding variables and research gaps with this
approach and provide a medical perspective on the use of biomarkers along with currently conventional diagnostic approach for formulating medical treatment decisions.
2.0 ORGAN AND TISSUE SYSTEMS
2.1 Salivary Glands
The major salivary glands are the parotid, submandibular, submaxillary, and sublingual glands. Besides these glands, there are many tiny glands called minor salivary glands located in the lips, inner cheek area (buccal
mucosa), and extensively in other linings of the mouth and throat. The epithelial cells of the salivary gland divide only rarely, hence this tissue would be expected to be relatively radiation resistant. In man, however,
the salivary gland shows a high sensitivity to ionizing radiation. The parotid gland seems to be more sensitive to irradiation than the submandibular gland [Henriksson et al. 1994], although the molecular
mechanism is not known. The post-irradiation induced proliferative activity was greater in the intercalated duct compartment of the parotid gland than that of the submandibular gland, which may be related to the
increased radiosensitivity [Peter et al. 1994]. Nagler suggested that the causes for the specific parotid radiosensitivity are transition, highly redox-active metal ions, such as Fe and Cu, associated with secretion
granules [Nagler et al. 1997].
A few hours after irradiation injury, cells in the salivary gland show acute inflammation and degenerative changes resulting in increases in plasma or serum amylase activity. An increase in serum amylase activity
(hyperamylasemia) from the irradiation of salivary tissue has been proposed as a biochemical measure of early radiation effect in a normal tissue [Becciolini et al. 1984; Leslie and Dische, 1992]. These concepts are based
on studies involving radio-iodine therapy [Maier and Bihl, 1987; Becciolini et al. 1994a, 1994b], radiation therapy [Chen et al. 1973; Dubray et al. 1992; Leslie and Dische, 1992; Becciolini et al. 2001], and recently a
radiation accident [Akashi et al. 2001]. Histochemical, isozyme analysis, and partial-body exposure studies confirm that the increase in serum amylase activity originates from the parotid glands.
Table 1: Candidate radiation biomarkers and functional tests from various tissue system or organs

Tissue system or organ Candidate radiation biomarker Candidate radiation bioindicator or functional test Radiation pathology Reference Gastrointestinal (GI) or Digestive System
Parotid salivary gland Amylase activity ↑ Serum or urinary amylase activity Mucositis Chen et al. 1973; Hofmann et al. 1990;
Dubray et al. 1992; Becciolini et al.


2001; Blakely et al. 2007; Blakely et al.
2010
Small
intestine
Citrulline, neurotensin and
gastrin hormones
↓ Serum or plasma citrulline,
neurotensin or gastrin;
↑ sugar concentration ratios using
dual-sugar permeability test
measured in serum
GI ARS
subsyndrome
Lutgens et al. 2003, 2004; Vigneulle et
al. 2002; Dublineau et al. 2004;
Bertho et al. 2008
Liver
C-reactive protein (CRP);
Serum amyloid A (SAA)
Oxysterol 7a-
hydroxycholesterol
↑ Serum or plasma CRP or SAA;
↑ Plasma oxysterol 7a-
hydroxycholesterol
ARS subsyndrome;
Hepatic tissue
radiation injury
Mal’tsev et al. 1978, 2006; Goltry et al.
1998; Koc et al. 2003; Roy et al. 2005;
Ossetrova et al. 2007; 2010;
Ossetrova and Blakely 2009; Blakely
et al. 2010;
Hemopoietic System
Bone
marrow
Flt-3 ligand (Ftl-3), IL-6, G-CSF ↑ Serum or plasma Flt-3
Bone marrow ARS
subsyndrome
Bertho et al. 2001, 2008
Cutaneous System

Cytokines (IL-1, IL-6, tumor
necrosis factor, GM-CSF,
TGF-β, intracellular adhesion
molecule, MMP
↑ IL-1, IL-6, GM-CSF, TGF- β,
intracellular adhesion molecule, and
MMP measured from skin tissues
Cutaneous ARS
subsyndrome
Martin et al. 1997; Ulrich et al. 2003;
Liu et al. 2006; Muller and Meineke
2007; Guipard et al. 2007
Respiratory System
Lung Oxysterol 27-hydrocholesterol
↑ plasma oxysterol 27-
hydrocholesterol
Respiratory ARS
subsyndrome
Roy et al. 2005
Cerebrovascular/Central Nervous System

Oxysteril 24S-
hydroxycholesterol
↑ plasma oxysteril 24S-
hydroxycholesterol
Cerebrovascular
ARS subyndrome
Roy et al. 2005


Figure 1: Radiation Biomarker Concept?Time Course. Schematic illustrating the time dependency of tissue- or organ-specific radiation biomarkers’ radioresponse. Representative data for parotid glands
[Chen et al., 1973], liver [Mal’tsev et al. 1978], GI [Lutgens et al. 2003], bone marrow [Bertho et al. 2001], and skin [Guipaud et al. 2007] are shown. See text for additional details.

Figure 2: Radiation Protein Biomarker Concept?Dose Response. Schematic illustrating the dose dependence of tissue- or organ-specific candidate radiation biomarkers’ radioresponse. Representative
data for parotid glands [Dubray et al., 1992], liver [Mal’tsev et al. 1978], GI [Lutgens et al. 2003], bone marrow [Bertho et al. 2001], and skin [Guipaud et al. 2007] are shown. See text for additional details.

Table 2: Select list of radiation-responsive blood based proteomic, metabolomic, and hematology biomarkers showing their dose range
(for various models) and time-window for meaningful diagnosis of radiation injury and dose*
Proposed blood or
serum biomarker
Pathways
Dose range (Gy) Time
window for
meaningful
diagnostics
References
Rodent
studies
NHP
studies
Human
radiation
therapy
Human
radiation
accidents
Salivary α-amylase
activity
Parotid gland
tissue injury
NA 0?8.5 Gy 0.5?10 Gy
3.5, 8, and
18 Gy
(Tokai-
mura)
12?36 h;
peaks at 24 h
Hofmann et al. 1990; Dubray
et al. 1992; Becciolini et al.
2001; Blakely et al. 2007;
Blakely et al. 2010
IL-6, G-CSF
Immunostimulatory
effects on bone
marrow cells
1?7 Gy 6.5 Gy NA 1?10 Gy
4?48 h;
3?8 d
Beetz et al. 1997; Gartel et al.
2002; Bellido et al. 1998;
Ossetrova et al. 2007, 2009
Flt-3 ligand
Bone marrow
aplasia
1?7 Gy 1?14 Gy NA
0.25 to 4.5
Gy
24 h?10 d
Bertho et al. 2001; Bertho et
al. 2008
CRP, SAA
Acute-phase
reaction
1?7 Gy
(SAA)
1?14 Gy
(CRP)
1?20 Gy
(CRP)
1?10 Gy
(CRP)
6 h?4 d;
5?14 d
Mal’tsev et al. 1978, 2006;
Koc et al. 2003; Goltry et al.
1998; Ossetrova et al. 2007,
2010; Ossetrova and Blakely,
2009; Blakely et al. 2010
Citrulline
Small bowel
epithelial injury
1?14 Gy Not done
1?20 Gy
(2-Gy daily
fractions)
~4.5 Gy >24 h
Lutgens et al. 2003, 2004;
Bertho et al. 2008
Lymphocytes,
neutrophils, and ratio
of neutrophils to
lymphoyctes
Hematopoietic
tissue injury
1?7 Gy 1?8.5 Gy 1?20 Gy 0?30 Gy 2 h?8 d
Goans et al. 1997; Guskova
et al. 1997; Blakely et al.
2005, 2007; Ossetrova et al.
2010
*Concept to use of multiple biomarkers for radiation injury and dose assessment (Blakely, Ossetrova et al., U.S. Patent Application No. 60/812,596.)


Serum amylase activity increases occur early after head and neck irradiation of humans [Kashima et al., 1965] and generally show peak values between 18?30 h after exposure, returning to normal levels within a
few days [Chen et al. 1973] (Figure 1). Radiation dose-dependent increases in the early (1 day) hyper-amylasemia are supported by radio-iodine therapy [Becciolini et al. 1994a, 1994b], radiotherapy [Hennequin
et al. 1989; Hofmann et al. 1990; Dubray et al. 1992; Becciolini et al. 2001], and limited data from three individuals exposed in a criticality accident [Akashi et al. 2001]. Significant inter-individual variations are
reported in these radiation studies [Chen et al. 1973; Dubray et al. 1992; Leslie and Dische, 1992] (Figure 2).
This inter-individual variation in biochemical response is not unexpected, since it is well known that the radiation level causing irreversible failure of the hematopoietic system varies among individuals and may
reflect genetic and physiological differences and relative differences in the radiosensitivity of hematopoietic stem/progenitor cells [Dainiak, 2002] as well as radiation exposure parameters (i.e., partial-body exposures,
shielding, dose-rate, etc.) [Koenig et al. 2005].
Limited studies have previously evaluated serum amylase activity radioresponse using rhesus-monkey radiation models. Dubray and colleagues cite an unpublished observation by L.C. Stephens demonstrating
radiation-induced increases in serum amylase activity using a rhesus monkey radiation model but note ―tremendous individual variation and no dose response relationship? apparent [Dubray et al. 1992]. Blakely
and colleagues [Blakely et al. 2007] using a rhesus monkey model system also reported significant inter-individual variation (3.4?to 30.5-fold at 1 day after irradiation) following exposure to whole-body acute
radiation exposure (6.5-Gy
60
Co- rays). Similar but less pronounced inter-individual variations (1.8?5.6 fold) were seen in the plasma amylase protein levels [Ossetrova et al. 2007].
2.2 Gastrointestinal system
There have been no survivors of those victims known to have been exposed to ionizing radiation doses of sufficient strength to cause injury to the gastrointestinal (GI) system [Monti et al. 2005; Genyao and
Changlin, 2005]. Although one approach of medicinal science has been to develop and evaluate treatments for this injury, another has been an attempt to identify a biomarker for this injury. Finding such a biomarker,
especially if it is expressed before GI injury is clinically evident, could assist medical personnel in triage of patients.
As reviewed by Lutgens and Lambin [Lutgens and Lambin, 2007], the current and most promising candidate biomarker for GI injury is the amino acid citrulline. Lutgens and colleagues showed that in mice following
total-body-irradiation (TBI) there was a radiation time- (Figure 1) and dose- (Figure 2) dependent decrease in plasma citrulline levels with a statistically significant radiation-dose citrulline-response relationship occur-
ring at 84-h post-irradiation [Lutgens et al. 2003]. At this time interval post-irradiation, the citrullinemia significantly correlated with both jejunal crypt regeneration and the measured circumference of the epithelial
surface lining. They also measured citrulline in patients undergoing abdominal fractionated radiotherapy and found significant decreases as a function of total radiation dose, the volume of bowel treated, and the clinical
toxicity grading [Lutgens et al. 2004].
The notion of citrulline as a GI marker for the status of intestinal enterocyte mass is based on several physiological principles. First, circulating citrulline is almost completely produced in the enterocytes of the
small intestine. Second, citrulline passes from the intestine via the portal hepatic vein to the liver. However, it is not metabolized in the liver, but from there passes into the general circulation to reach the kidney, where
it is metabolized to arginine in the proximal renal tubules (Figure 3).

Figure 3: Serum citrulline as a potential surrogate marker for radiation-induced intestinal damage. Schematic illustrates the mechanisms for radiation induced decreases in plasma citrulline, derived
from the intestinal-hepato-renal axis. See manuscript text for additional details.
These principles were first taken advantage of by Crenn and colleagues, who showed a significant decrease in plasma levels of citrulline associated with the level of bowel failure in 57 patients, who had undergone
bowel resection (e.g., short bowel syndrome) at least two years earlier [Crenn et al. 2000]. In that study, citrulline concentration was measured along with parenteral nutrition dependence to define permanent and
transient intestinal failure. Citrulline levels were significantly lower in the patients than in the control population, and the citrullinemia was significantly correlated with bowel length. Citrulline has also been
shown to be reduced in patients, who had undergone small-intestinal transplantation and in which graft had begun to show signs of rejection [David et al. 2006, 2007].
There are three other promising biomarkers for radiation-induced GI damage. The first is a clinical dual sugar permeability test. This test has been used successfully in patients with several GI disorders, for example, celiac
disease [Cox et al. 1999]. In this test, a solution of two inert and non-absorbable sugars of unequal diameters and in equal concentrations was taken orally. Lactulose and rhamnose were two such examples, the former
being a disaccharide, the latter, a monosaccharide (although other inert agents can be used). Two were used because they both were affected in the same manner by GI transit, gastric emptying, etc.
The sugars passively cross the GI mucosa, the larger one by the paracellular pathway (e.g., tight junctions), and the smaller one by the transcellular pathway?or through the cell membrane?although there are several other
hypotheses as to how this transport occurs under normal conditions. Transport of the smaller sugar prevails and the ratio of the two recovered concentrations, for example lactulose/rhamnose (L/R), under normal conditions,
favors the smaller sugar and the value is small. Under pathological conditions, the reverse occurs and passage of the larger molecule dominates the transport processes, increasing the value of the L/R ratio.
Historically, the ratio of these two sugars was measured from urine, 5?6 h after ingestion. Recently however, it has been shown that the ratio can be recovered from the serum within 1?2 hours after ingestion of the
sugars [Cox et al. 1999]. The serum values within that short time frame are very similar to the values recovered in urine after 6 h. This time savings would greatly facilitate any medical management for someone
who might have a GI radiation injury. A new, dried blood spot (DBS) technology has been applied to this sugar permeability testing, which requires only a pin-prick on the finger to retrieve enough blood (~ 25 μl)
for analysis [Katouzian et al. 2005]. In addition, the DBS methodology has been used for monitoring serum citrulline values and there is a strong linear correlation between DBS-reported citrulline concentrations and
those from HPLC [Yu et al. 2005].
While there have been some clinical evaluations of intestinal permeability following irradiation, these evaluations have been performed and associated with patients undergoing fractionated radiotherapy [Pia de
la Maza et al. 2001]. In addition, there has been one instance in which the sugar permeability test has been used experimentally in the nonhuman primate [Vigneulle et al. 2002]. In this study, the ratios of serum levels
of lactulose vs. 3-O-methylglucose (3-OM) and lactulose vs. mannitol were respectively measured to eval-uate the respective transcellular and paracellular permeabilities before and at several time intervals after 9.5-
Gy total abdominal irradiation (TAI) in the nonhuman primates. They found a statistically significant changes (i.e., increase in permeabilities for both sugars) at 7-days post-irradiation, although there were non-
significant increases on d 5 and days beyond 7 d. Further studies are needed in this area to make this approach useful for diagnostic applications.
The other two potential markers are the GI hormones gastrin and neurotensin (NT). Dublineau and colleagues investigated a panel of 7 hormones following 16-Gy TBI in pigs [Dublineau et al. 2004]. Of these
GI hormones, only gastrin and NT dramatically changed within 24-h post-irradiation, both falling. The former hormonal change was thought to reflect stomach damage; the latter, small intestine. Although the NT
data from this study are more compelling, the small sample size (n = 3) of the study strongly suggest further verification of these observations.
2.3 Hematopoietic (bone marrow) system
Bertho and colleagues proposed using animal irradiation models that plasma flt-3 ligand is a potential new bioindicator for radiation-induced aplasia (Figures 1 and 2) [Bertho et al. 2001]. Plasma flt-3 ligand concen-
tration also was correlated with radiation-induced bone marrow damage using local fractionated radiotherapy study [Hutchet et al. 2003]. The number of circulating white blood cells and platelets inversely correlated with plasma flt-3 ligand levels. In a recent radiation accident the measured flt-3 ligand levels were indicative
of the severity of bone marrow aplasia [Bertho et al. 2008; Bertho and Roy, 2009].
2.4 Hepatic (liver) system
C-reactive protein (CRP) is primarily formed in the liver and an acknowledged non-specific biomarker for various stresses. Plasma CRP increases is an exquisitely sensitive systemic marker of inflammation and
tissue damage and also has been shown to play an essential role in radiation injury [Tukachinski and Moiseeva, 1961; Mal’tsev et al. 1978, 2006; Ossetrov et al. 2007; Blakely et al. 2010]. Time- (Figure 1) and
dose-response (Figure 2) calibration curves for CRP expression measured in blood of 70 nonhuman primates -irradiated to a broad dose range up to 12 Gy and time-points from 2 h to 30 d revealed significantly
increased plasma CRP levels observed at 8?72 h post irradiation with a threshold about 0.5 Gy [Mal’tsev et al. 1978]. CRP levels were determined using a method of capillary precipitation of specific C-reactive
antiserum (the most sensitive existing method at that time). The time-interval for the second phase of appearance of CRP in the blood of irradiated animals correlate with the time interval for the expressed development of the cytolytic and destructive processes induced by irradiation [Mal’tsev et al. 1978].
Numerous studies show that dynamics and content of CRP exactly reflect the course and severity of the radiation sickness and may play a role as a factor in the prognosis [Petrov, 1962; Tukachinski and Moiseeva,
1961]. Mal’tsev and colleagues reported that indexes of CRP content in a peripheral blood of 147 patients damaged at the Chernobyl accident have been found to provide information for the prognosis of the probable
level of acute radiation sickness [Mal’tsev et al. 2006].
Roy and colleagues have proposed that the plasma concentration of the oxysterol 7α-hydroxycholesterol reflects hepatic damage following irradiation [Roy et al. 2005]. This oxysterol results from liver tissue
specific enzymatic degradation (via cytochrome P450) of cholesterol (CYP7A1). In this report, after a 10-Gy TBI to rats, there was a five-fold and statistically significant decrease in this specific sterol on the third day
post-irradiation when compared with controls. Roy and co-workers suggest that its diminished levels are part of multi-organ radiation damage.
2.5 Respiratory system
While increases in plasma levels of the oxysterol 27-hydroxycholesterol are reported to reflect pulmonary damage, Roy and colleagues have found that the plasma concentration of the 2,7-hydroxycholesterol is
unchanged following irradiation [Roy et al. 2005]. This oxysterol results from lung tissue specific enzymatic degradation (via cytochrome P450) of cholesterol (CYP27A1). These authors suggest that these levels
remained within control values because other undamaged tissues also produce and release it into the circulation.
2.6 Cerebrovascular/central nervous system
Roy and colleagues have proposed that increases in the plasma concentration of the oxysterol 2,4 S-hydroxycholesterol reflects brain damage following irradiation [Roy et al. 2005]. The oxysterol results from
brain tissue specific enzymatic degradation (via cytochrome P450) of cholesterol (CYP46A1). In this report, after a 10-Gy TBI to rats there was a statistically significant increase in this specific sterol on the third day
post-irradiation. As discussed above, Roy and co-workers suggest that these elevated levels of oxysterols are indicative of multi-organ radiation damage.
2.7 Cutaneous system
The symptoms of the cutaneous radiation syndrome (CRS) are based on a combination of inflammatory processes and altered cellular proliferation, all of which result from a specific pattern of transcription-activated
pro-inflammatory cytokines and growth factors. In the simplest terms, the phases can be distinguished as the prodromal stage, the manifest illness stage, and the chronic stage [Meineke, 2005], although [Peter et al. 2001]
have further subdivided and defined them by their latency and persistence. In this latter scheme, the latency of the prodromal stage is within minutes to hours, and can persist from 0.5 to 36 hours. The manifestation stage
has a latency of 3 weeks, and can last for 1?2 weeks. The former stage is manifest by erythema and pruritis; the latter, by these symptoms as well as bullae and ulcers. The prodromal stage typically occurs after 2-Gy
irradiation, while the other stages occur after radiation doses greater than 3 Gy.
1

Muller and Meineke [Muller and Meineke, 2007] reported that cutaneous tissue’s radioresponse involves the major cytokines including changes in interleukin-1 (IL-1) in both forms (IL-1α and IL-1β), IL-6, tumor
necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β), as well as granulocyte-macrophage colony-stimulating factor (GM-CSF), and such chemotactic cytokines, as IL-8 and eotaxin. This is a

1
Personal communication with Dr. Viktor Meineke, Bundeswehr Institute of Radiobiology, Munich, Germany. burgeoning new area of work and much of the data are from in vitro research. From those data taken from
skin or skin biopsies, the following items are noteworthy for this report. In skin taken from mice, Liu and colleagues have shown increased expression of IL-1β and matrix metalloproteases (MMPs) at 19 d after 30-
Gy irradiation [Liu et al. 2006]. This was the time when early dermatitis appeared. In skin taken from pig 6-h after a 16-Gy irradiation, Martin showed significantly elevated TGF-1β gene expression although this was
transient, returning to control values at 24 h [Martin et al. 1997]. Whether and how this is related to the involvement of TGF-1β with late development of radiation-induced skin lesions is unknown. In a study of
skin biopsies from patients presenting with basal cell carcinoma, Muller and colleagues found elevated levels of intercellular adhesion molecule-1 (ICAM-1) after a 15-Gy cumulative radiotherapy treatment [Muller et
al. 2006]. These results were compared with biopsy material taken before irradiation. Since the tissue was taken from the tumor site, these results may not reflect normal tissue. Lastly, and related to the elevated
MMP levels, Ulrich and colleagues have demonstrated that serum levels of MMP-2 and MMP-9 are signify-cantly elevated between 3 and 14 d in patients with burns, who underwent skin excision and autografting
[Ulrich et al. 2003]. These patients’ cases were compared with a control group undergoing elective plastic surgery. MMPs are zinc-dependent endopeptidases in general and MMP-2 and MMP-9 are implicated in
tissue maintenance/repair and are elevated after thermal injury.
Using a novel approach to irradiate only the skin in a murine model, Guipaud and colleagues recently evaluated 64 serum proteins following varied radiation doses (Figures 1 and 2). The analysis was done on
days 1, 5, 14, 21, and 33 post-irradiation [Guipaud et al. 2007]. While the greatest level of expression (whether up or down) was seen on day 14, there were a number of proteins significantly different from
control in days 1 and 5. Many of these were acute-phase proteins, associated with injury of any sort, while some are involved in coagulation.
In summary, while there are numerous clinical indices and novel medical devices used to score the CRS [Peter et al. 2001], the notion of biomarkers for such an injury and the research in this area are in its infancy.
3. CONFOUNDING VARIABLES, GAPS, AND LIMITATIONS
Severe inflammation of the salivary glands, pancreas, and gastrointestinal tissues can cause increases in serum alpha-amylase activity. For example, 10-fold or greater elevations can be indicative of pancreatitis, cancer of
the pancreas, gall-bladder disease, and mumps. Five- to ten-fold increases may indicate renal failure and disease of gastrointestinal tissue as well as salivary gland trauma. The time course for the radioresponse
elevations of serum alpha-amylase is from 18 to 36 h after irradiation, with a peak value at 24 h and returning to near control levels by 48 h (Figure 1). The transitory elevation in serum alpha-amylase activity, while
limiting the diagnostic utility of this radiation biomarker for practical applications, can be useful to rule out non-radiation pathologies.
Hematopoietic cytokines are involved in the proliferation and differentiation of various blood cell progenitor cell populations. Flt-3 ligand stimulates various blood cell populations, including neutrophils. Disorders that
result in the induction of proliferation of hematopoietic progenitor cell populations would be expected to cause elevations in plasma hematopoietic cytokines. In addition, hematopoietic cytokines are elevated during
the initial acute-phase of inflammation, particularly as a result of bacterial infection and some cancers.
However, normal peripheral blood neutrophil counts, along with an expected corresponding lower baseline serum Flt-3 levels, are seen in certain populations (i.e., people of African and Middle Eastern descent)
[Bertho et al. 2008; Bertho and Roy, 2009]. Diagnostic use of serum hematopoietic cytokines for radiation exposure assessment will require comparison of results with baseline levels appropriate controls.
CRP is one member of the acute-phase reactants and increases dramatically (>100-fold) during the inflame-mation process and is believed to play a role in innate immunity, as an early defense against infections.
Moderate increases in CRP are associated with increased risk of diabetes, hypertension, and cardiovascular disease. Normal concentration in healthy human serum is usually lower than 10 mg/L, slightly increasing with
ageing. Higher levels are found in late pregnant women, mild inflammation and viral infections (10?40 mg/L), active inflammation, bacterial infection (40?200 mg/L), severe bacterial infections and burns (>200 mg/L)
[Clyne and Olshaker 1999]. It might be elevated with complications or treatment failures in patients with pneumonia, pancreatitis, pelvic inflammatory disease (PID), and urinary tract infections. In patients with
meningitis, neonatal sepsis, and occult bacteremia, CRP is also usually elevated. As a generally acknowledged non-specific biomarker for a variety of disorders, elevated CRP levels cannot be used alone for a definitive
specific diagnosis. AFRRI scientists have recently advocated using elevated CRP levels as an early-phase triage tool to identify individuals suspected of severe life-threatening radiation exposure [Ossetrova et al. 2007;
Blakely et al. 2010]. This concept is based on Mal’tsev and colleagues’ results [Mal’tsev et al. 1978], which show that CRP levels increase after radiation exposure based on dose- and time-radioresponse studies using a
nonhuman primate model, and results from analysis of samples from Chernobyl victims (Figures 1 and 2) [Mal’tsev et al. 2006]. Plasma CRP levels at 1 to 3 d after radiation doses greater than 1 Gy, based on the
nonhuman primate radiation model of Mal’tsev, are significantly higher from baseline level.
With regard to GI markers for radiation damage, there are numerous gaps in the knowledge and under-standing of the potential markers discussed. First and foremost, each of these markers must be repeatedly
tested and validated in other animal models. Citrulline, for example, should be evaluated in an animal model that would allow for serial sampling, like what was done with the dual-sugar permeability test and the panel
of gut hormones (e.g., gastrin and NT).
In addition, there should be investigations of the dose-response characteristics of each potential biomarker. This was best done for citrulline, but the other markers were evaluated only after a single isolated dose of
ionizing radiation. It will be important to know how the other markers compare with regard to their dose-responsiveness to lower doses of irradiation. This may be especially true for gastrin and NT, since the pig is
not a well-characterized animal model for studying the GI syndrome. The benchmark for the GI syndrome has always been the radiation dose corresponding to the LD50/6 or LD50/7, a value that is more readily
attainable in rodents.
A third limitation of these biomarkers is with regard to the timing of its appearance. Lutgens and colleagues reported in mice after single dose TBI a significant dose-response relationship for radiation vs. citrulline only
84-h post-irradiation [Lutgens et al. 2003]. If this time interval were identical in humans, they might already be showing signs and symptoms (i.e., diarrhea, etc.) of the GI radiation syndrome. Evaluation of such signs
and symptoms in rodents is not easy to accomplish. In addition, post-irradiation diarrhea in the rodent can be very transient. With regard to the intestinal permeability testing, it is not known whether such a clinical test
could predict the onset of diarrhea, for example, or intestinal damage. Vigneulle and colleagues reported the onset of diarrhea five d following 9.5-Gy TAI [Vigneulle et al. 2002]. While the L/3-OM ratio was elevated
on this day, both this ratio and the L/M ratio were not significantly elevated until seven days post-irradiation, after the onset of diarrhea. Other than prior to irradiation to determine the control values, the testing was not
done before day 5 post-irradiation. The authors also report that the diarrhea continued through 14 d post-irradiation but the L/3-OM ratio did not return to normal until 27 d post-irradiation and the L/M ratio did not
return to normal until 35 d post-irradiation. The falls in gastrin and NT reported by Dublineau and colleagues were 24 h post-irradiation [Dublineau et al. 2004]. It may be that the rate of change in one of these markers
at early time intervals post-irradiation can be used for predicting that organ damage, rather than the precise value. The data from mice on citrulline clearly show that different rates of fall over the first several days post-irradiation are radiation dose-dependent.
For the individual specific biomarkers described (e.g., citrulline, gastrin, and NT), there may be potentially confounding variables that need to be evaluated in order to discriminate GI organ damage or pathology from
an insult other than irradiation. For example, it has been shown in the nonhuman primate that prolonged fasting can lower serum citrulline concentrations [Cameron et al. 1985]. The clinical literature has not been
so precise. In one study, Beaumier and colleagues showed that serum citrulline values in humans were significantly greater after a meal but these differences disappeared if the meal was supplemented with L-
arginine [Beaumier et al. 1995]. Conversely, this same group showed no difference in serum citrulline values between the fed and fasted states under similar conditions [Castillo et al. 1995]. There has been one clinical
report that intestinal permeability is increased during malnutrition but this is somewhat controversial [Ferraris and Carey, 2000] and the data are limited. George and colleagues showed that fasting and a
circadian pattern of food intake can influence circulating levels of NT [George et al. 1987]. Levels of gastrin also fall during starvation [Lichtenberger et al. 1976; Goodlad et al. 1983]. Because it is well documented
that food and water intake are reduced following ionizing radiation, and in a radiation-dose-dependent manner, it may be important to understand how this alone contributes to the reduced citrulline, gastrin or NT
observed following radiation.
4. MEDICAL PERSPECTIVE ON USE OF BIOMARKERS ALONG WITH CURRENT RECOMMENDED METREPOL ARS SEVERITY SCORE
INDICATORS
Medical treatment decisions of radiation victims were traditionally based on assessment and reconstruction of the radiation dose applied to the individual. Physical parameter-based classification systems have some
major drawbacks for clinicians treating radiation victims. The main interest of clinicians is applying medical care to improve patients’ outcome. Therefore, a severely score based on a classification system and/or bio-
markers levels should ideally predict the outcome of a patient, taking into account the kind of radiation including radiation quality, the heterogeneity and the individual radiation sensitivity. Furthermore, con-
comitant injuries or disorders have a major impact on the clinical course of the irradiated person. The ideal classification marker should integrate all this topics and the application of the marker should be easy, fast
and worldwide accessible. To date only CRP levels have been evaluated as a biomarker or classification system to predict the outcome of irradiated persons (Mal’tsev et al. 2006).
In medical settings, a lot of data can be gained easily by the physicians and their routine applications. For instance, medical history-taking, medical examination, and basic laboratory testing are accessible at any site
where radiation victims can be treated. Fliedner and co-workers spent enormous efforts in creating a database called SEARCH (System for Evaluation and Archiving of Radiation accidents based on Case Histories). Based
on the database, which contains radiation accidents and medical recordings over the last 50 years, an evidence-based clinical classification system was developed [Fliedner et al. 2001]. The concept of the ―response
categories? (RC) focuses not on the etiology of the radiation syndrome (physical dose, biological dose) but on the changes in the health status of the individual. Using easy-to-acquire clinical signs and focusing on the
organ-specific changes, the most important organ-system alterations were used for grading to create an overall classifier called response category. The organ systems used are neurovascular, hematopoietic, cutaneous and
gastrointestinal. Every organ system is graded from 1 to 4 and the integration is the RC.
As an example, the hematopoietic system is described briefly. Impairment of hematopoiesis by whole-body irradiation leads to clinical symptoms like susceptibility to infections, bleeding disorders or wound healing
disorders, which are summarized under the term hematopoietic syndrome (HS) of the acute radiation syn-drome. The radiosensitivity of hematopoietic stem cells and their damage in the bone marrow is the patho-
physiologic background of the hematopoietic syndrome. The change of lymphocyte, granulocyte and platelet counts over time can be used for a clinical-based classification system. The repeated measurement of single
parameters is essential for the grading of the hematopoietic syndrome. Irradiation leads to hypoplasia or aplasia of the bone marrow, resulting in pancytopenia. Although the probability of the HS increases with
physical dose applied, there is no save threshold. Usually the HS occurs at total-body doses from 1 to 1.5 Gy, but the dose-effect concept is of little importance for the clinical grading of individuals [Fliedner et al. 2001;
Friesecke et al. 2000]. A H4 (hematopoietic syndrome grade 4) damage shows a rapid decline for lympho-cytes within 24 h below 0.25 × 109/L, an initial granulocytosis up to 48 hours followed by a rapid decline
below 0.5 × 109/L. The nadir reached for lymphocytes, granulocytes and thrombocytes will last for at least several weeks. Patients graded as H3 show a decline in lymphocytes within the first 48 h down to 0.25 ×
109/L and 1.0 × 109/L. There is also an initial granulocytosis followed by a subsequent decrease until day 5.
A very important finding is an abortive rise starting at around day 5, increasing granulocyte counts again for about 5?8 days and followed by a second decline down to 0.5 × 109
/L. The nadir of the platelets will be reached with counts from 0?50 × 109/L around day 16?18. Autologous recovery will start around day 30?40. In H2 patients lymphocytes usually stay between 0.5 × 109
/L and 1.5 × 109L, granulocytes drop below 1.0 × 109/L around day 20 and thrombocytes show a nadir of around 50 × 109/L cells.
This description of the HS grading is a vast simplification of the grading process. The curve of the cell changes, including abortive rise, are the basis for the grading and their interpretation needs expert
knowledge. Patients with RC1 need little support to cope with the radiation damage. In RC2 patients, autologous recovery is certain and medical treatments are needed to bridge transient damage. RC3 patients
need maximum medical effort to be rescued including, for instance, differential antimicrobial therapy, cytokines or reverse isolation. When the radiation damage is graded as RC4, irreversible organ damage will
occur thus leading to multiple organ failure. There is a small therapeutic window for patients with a grade-4 hematopoietic syndrome (H4) to be rescued by stem-cell transplantation. The detailed grading system was
published in 2001 [Fliedner et al. 2001] and adoptions from scientific societies and groups were widespread widely spread [Dainiak et al. 2003; Waselenko et al. 2004; Gorin et al. 2006; Ganser 2007]. The RC concept
allows predicting the clinical course of the HS and the probability of autochtone regeneration [Fliedner et al. 2007]. Interpretation difficulties may arise in combined injuries accompanying diseases, such as in patients
with low exposition of the ARS severity level or in early diagnostic situations. Only patients with a very high probability to develop multiple organ failure due to very high radiation express clinical signs and symptoms
to permit clinical classification systems to predict the clinical course in the first hours. Granulocyte and lymphocyte kinetics or their ratio might offer the potential for early diagnosis but evaluation in radiation
accidents is still a matter of research.
Biomarkers offer a great opportunity to solve many remaining problems. For example, Mal’tsev and colleagues show that early-phase (1?2 and 3?9 d) CRP levels measured in Chernobyl victims was correlated
with the ARS sub-syndrome severity levels (Mal’tsev et al. 2006). Similar studies need to be performed for additional candidate organ-specific biomarkers. The development and evaluation of biomarkers uses cell
culture experiments, animal models or medical treatment procedures like radiation therapy or nuclear medicine applications. The advantage of these models is the simple determination or measurement of applied
physical dose, which then can be used easily to build dose effect curves. The ―gold standard? biomarker is the lymphocyte dicentric bioassay introduced in 1962 [Bender and Gooch, 1962]. Although robust and
widely used, its application is time consuming and laborious. Evaluation of measurements in radiation accidents and correlation to the response categories of patients show a concordance of about 70?80% of
measurements, depending on the cut off for RC4 patients, although distinguishing between RC2 and RC3 patients was not possible in the cohort examined.
2

An ideal radiation injury biomarker would include fast and reliable measurements, the potential for automation of the measurements, small and easy to obtain sample specimens, and the use of standard
detection technology, obviating the need for special training. Also, the biomarker should be correlated both to physical dose as well as ARS severity score levels, similar to that done by Mal’tsev and colleagues
[Mal’tsev et al. 2006], which would permit prediction of clinical outcome of future radiation scenarios such as radiation accidents. Medical applications like radiation therapy might help to test the markers in humans.
Today, many different biological, chemical and even physical effects are used to develop biological markers.
Our research focuses in changes of gene expression or protein measurements. Ongoing research demonstrates that mRNA changes in three genes, namely GADD45α, CDKN1 and ATF3, at 8, 24 and 48 h
post irradiation correlate with radiation dose from 0 to 2 Gy (data not shown). Similar findings were previously demonstrated by Amundson, Blakely, and Grace [Amundson et al. 2000; Blakely et al. 2003;
Grace et al. 2002; Amundson et al. 2004; Grace and Blakely 2007]. Use of multiple biomarkers today offer the opportunity to verify that irradiation has happened and to predict the outcome of a group of irradiated
patients with a distinct probability, which might help to plan and effectively use the available resources in a mass-casualty situation.
There is a clear need to develop and prospectively test biomarkers for radiation casualties. This is important not only for prediction of clinical outcome and assistance in clinical decision-making, but also for under-
standing acute radiation syndrome and developing new medical-care strategies. The authors strongly believe that only a combined approach with a set of distinct genes, proteins or other biomarkers in combination with
clinical classification systems might improve the rapid and correct classification of radiation victims.
5. ACKNOWLEDGMENTS
The views expressed here are those of the authors; no endorsement by the U.S. Department of Defense has been given or inferred. AFRRI supported this research under work units RAB4AL, RAB4AM, and
RBB4AR. The authors wish to thank David J. Sandgren for his assistance and AFRRI’s editorial staff for their expert editorial contributions. and AFRRI’s editorial staff for their expert editorial contribution.
6. REFERENCES


03. 2012年7月17日 01:52:57 : pi0fYycp4s
[1] Akashi, M., Hirama, T., Tanosaki, S., Kuroiwa, N., Nakagawa, K., Tsuji, H., Kato, H., Yamada, S., Kamata, T., Kinugasa, T., Ariga, H., Maekawa, K., Suzuki, G., and

Tsujii, H., Initial symptoms of acute radiation syndrome in the JCO criticality accident in Tokai-mura. Journal of Radiation Research (Tokyo) 42 Suppl.:S157?66, (2001).
[2] Amundson, S.A., Do, K.T., Shahab, S., Bittner, M., Meltzer, P., Trent, J., and Fornace, Jr., A.J., 2000. Identification of potential mRNA biomarkers in peripheral

blood lymphocytes for human exposure to ionizing radiation. Radiation Research 154:342?346, (2000).
[3] Amundson, S.A., Grace, M.B., McLeland, C.B., Epperly, M.W., Yeager, A., Zhan, Q., Greenberger, J.S., and Fornace, Jr., A.J., Human in vivo radiation-induced

biomarkers: gene expression changes in radiotherapy patients. Cancer Research 64:6368?6371, (2004).

2
Abend, M., Port, M., Dorr, H., Pieper, B., Fliedner, T.M., and V. Meineke, V., Correlation of radiation dose with clinical hematological response in radiation accident

victims, a retrospective study (in preparation).

[4] Beaumier, L., Castillo, L., Ajami, A.M., and Young, V.R., Urea cycle intermediate kinetics and nitrate excretion at normal and ―therapeutic? intakes of arginine in

humans. American Journal of Physiology 268 (Endocrinol. Metab.32):E884?E896, (1995).
[5] Becciolini, A., Giannardi, G., Cionini, L., Porciani, S., Fallai, C., and Pirtoli, L., Plasma amylase activity as biochemical indicator of radiation injury to salivary glands.

Acta Radiology [Oncology] 23:9?14, (1984).
[6] Becciolini, A., Porciani, S., Lanini, A., Benucci, A., Castagnoli, A. and Pupi, A., Serum amylase and tissue polypeptide antigen as biochemical indicator of salivary

gland injury during iodine-131 therapy. European Journal of Nuclear Medicine 21:1121?5, (1994a).
[7] Becciolini, A., Porciani, S., and Lanini, A., Marker determination for response monitoring: radio-therapy and disappearance curves. International Journal of Biological

Markers 9:38?42, (1994b).
[8] Becciolini, A., Porciani, S., Lanini, A., Balzi, M., and Faroani, P., Proposal for biochemical dosimeter for prolonged space flights. Physica Medica 17 Suppl. 1:185?6,

(2001).
[9] Beetz, A., Messer, G., Oppel, T., van Beuningen, D., Peter, R.U., Kind, P., Induction of interleukin 6 by ionizing radiation in a human epithelial cell line: Control by

corticosteroids. International Journal of Radiation Biology 72:3?43, (1997).
[10] Bellido, T., O’Brien, C.A., Roberson, P.K., Manolagas, S.C., Transcriptional activation of the p21 (WAF1, CIP1, SDI1) gene by interleukin-6 type cytokines: A

prerequisite for their pro-differentiating and anti-apoptotic effects on human osteoblastic cells. Journal of Biological Chemistry 273:21137?21144, (1998).
[11] Bender, M.A. and Gooch, P.C., Persistent chromosome aberrations in irradiated human subjects. Radiation Research 16:44?53, (1962).
[12] Bertho, J.M., Demarquay, C., Frick, J., Joubert, C., Srenales, S., Jacquet, N., Sorokine-Durm, I., Chau, Q., Lopez, M., Aigueperse, J., Gorin, N.C., and Gourmelon

P., Level of Flt3-ligand in plasma: a possible new bio-indicator for radiation-induced aplasia. International Journal of Radiation Biology 77: 703?12, (2001).
[13] Bertho, J.M., Roy, L., Souidi, M., Benderitter, M., Gueguen, Y., Lataillade, J.J., Prat, M., Fagot, T., De Revel, T., and Gourmelon, P. New biological indicators to

evaluate and monitor radiation-induced damage: an accident case report. Radiation Research 169:543?50, (2008).
[14] Bertho, J.M., and Roy, L., A rapid multiparametric method for victim triage in cases of accidental protracted irradiation or delayed analysis. British Journal of

Radiology 82:764?70, (2009).
[15] Bertho, J.M., Roy, L., Souidi, M., Benderitter, M., Bey, E., Racine, R., Fagot, T., Gourmelon, P., Initial evaluation and follow-up of acute radiation syndrome in two

patients from the Dakar accident. Biomarkers 14:94?102, (2009).
[16] Blakely, W.F., Miller, A.C., Grace, M.B., McLeland, C.B., Luo, L., Muderhwa, J.M., Miner, V.L., and Prasanna, P.G., Radiation biodosimetry: applications for

spaceflight. Advances in Space Research 31:1487?1493, (2003a).
[17] Blakely, W.F., Miller, A.C., Grace, M.B., McLeland, C.B., Muderhwa, J.M., Prasanna, P.G.S., Dose assessment based on molecular biomarkers. In: Radiation Safety

Aspects of Homeland Security and Emergency Response, Proceedings of the 36th Midyear Topical Meeting, Health Physics Society, McLean, VA, 229?234, (2003b).
[18] Blakely, W.F., Salter, C.A., and Prasanna, P.G., Early-response biological dosimetry?recommended countermeasure enhancements for mass-casualty radiological

incidents and terrorism. Health Physics 89(5):494?504, (2005).
[19] Blakely, W.F., N. I. Ossetrova, G. L. Manglapus, C. A. Salter, I. H. Levine, W. E. Jackson, M. B. Grace, P. G. S. Prasanna, D. J. Sandgren and G. D. Ledney. Amylase

and blood cell-count hema-tological radiation-injury biomarkers in a rhesus monkey radiation model?use of multiparameter and integrated biological dosimetry. Radiation

Measurements 42(6?7):1164?1170, (2007).
[20] Blakely, W.F., N. I. Ossetrova, M. H. Whitnall, D. J. Sandgren, V. I. Krivokrysenko, A. Shakhov, and E. Feinstein. Multiple parameter radiation injury assessment

using a nonhuman primate radiation model?biodosimetry applications. Health Physics 98:153?9, (2010).
[21] Cameron, J.L., Koerker, D.J., and Steiner, R.A., Metabolic changes during maturation of male monkeys: possible signals for onset of puberty. American Journal of

Physiology 249 (Endocrinology and Metabolism Clinics of North America 12):E385?E391, (1985).
[22] Castillo, L., Sanchez, M., Vogt, J., Chapman, T.E., DeRojas-Walker, T.C., Tannenbaum, S.R., Ajami, A.M., and Young, V.R., Plasma arginine, citrulline, and

ornithine kinetics in adults, with observations on nitric oxide synthesis. American Journal of Physiology 268 (Endocrinology and Metabolism Clinics of North America

31):E360?E367, (1995).
[23] Chen, W.; Kereiakes, J.G.; Silberstein, E.B., Aron, B.S., and Saenger, E. L., Radiation induces changes in serum and urinary amylase levels in man. Radiation

Research 54:141?151, (1973).
[24] Crenn, P., Coudray-Lucas, C., Cynober, L., and Messing, B., Post-absorptive plasma citrulline concentration: a marker of intestinal failure in humans. Transplant

Proceedings 30(6):2528, (1988).
[25] Crenn, P., Coudray-Lucas, C., Thuiller, F., Cynober, L., and Messing, B., Postabsorptive plasma citrulline concentration is a marker of absorptive enterocyte mass

and intestinal failure in humans. Gastroenterology 119:1496?1505, (2000).
[26] Cox, M.A., Lewis, K.O., and Cooper, B.T., Measurement of small intestinal permeability markers, lactulose and mannitol in serum. Results in Celiac Disease.

Digestive Diseases and Science 44:402?6, (1999).
[27] Clyne, B. and Olshaker J.S., The C-reactive protein. Journal of Emergency Medicine 17(6):1019?1025, (1999).
[28] Dainiak, N., Waselenko, J.K., Armitage, J.O., MacVittie, T.J., and Farese, A.M., The hematologist and radiation casualties. Hematology. American Society of

Hematology Education Program. 473?496, (2003).
[29] David, A.I., Gaynor, J.J., Zis, P.P., Conanan, L., Goldsmith, L., Esquenazi, V., Selvaggi, G. Weppler, D., Nishida, S., Moon, J., Madariaga, J.R., Ruiz, P., Kato T.,

Levi, D.M., Kleiner, G., Tryphonopoulos, P., and Tzakis, A.G.., An association of lower serum citrulline levels within 30 days of acute rejection in patients following small

intestine transplantation. Transplantation Proceedings 38:1731?2, (2006).
[30] David, A.I., Selvaggi, G., Ruiz, P., Gaynor J.J., Tryphonopoulos, P., Kleiner, G., Moon, J., Nishida, S., Pappas, P.A., Conanan, L., Weppler, D., Esquenazi, V.,

Levei, D.M., Kato, T., and Tzakis, A.G., Blood citrulline level is an exclusionary marker for significant acute rejection after intestinal transplantation. Transplantation

84:1077?81, (2007).
[31] Dublineau, I., Dudoignon, N., Monti, P., Combes, O., Wysocki, J., Grison, S., Baudelin, C., Griffiths, N.M., and Scanff, P., Screening of a large panel of

gastrointestinal peptide plasma levels is not adapted for the evaluation of digestive damage following irradiation. Canadian Journal of Physiology and Pharmacology

82:103?13, (2004).
[32] Dubray, B., Girinski, T., Thames, H.D., Becciolini, A., Porciani, S., Hennequin, C., Socie, G., Bonnay, M., Cosset, J.M., Post-irradiation hyperamylasemia as a

biological dosimetry. Radiotherapy Oncology 24(1), 21?6, (1992).
[33] Ferraris, R.P., and Cary, H.V., Intestinal transport during fasting and malnutrition. Annual Review of Nutrition 20:195?219, (2000).
[34] Fliedner T.M., I. Friesecke, and K. Beyrer. Medical management of radiation accidents. Manual on the acute radiation syndrome. The British Institute of Radiology,

London, United Kingdom, pp 1?66, (2001).
[35] Fliedner, T. M., Graessle, D., Meineke, V., and H. Dorr, H., Pathophysiological principles underlying the blood cell concentration responses used to assess the

severity of effect after accidental whole-body
radiation exposure: an essential basis for an evidence-based clinical triage. Experimental Hematology 35:8?16, (2007).
[36] Friesecke, I., Beyrer, K., Wedel, R., Reimers, K., and Fliedner, T. M., SEARCH: a system for evaluation and archiving of radiation accidents based on case histories.

Radiation Environmental Biophysics 39:213?217, (2000).
[37] Goltry, K.L., Epperly, M.W., Greenberger, J.S., Induction of serum amyloid A inflammatory response genes in irradiated bone marrow cells. Radiation Research

149:570?8, (1998).
[38] Ganser, A.A., Die hamatopoetische Form in Der Strahlenunfall. Veroffentlichungen der Strahlen-schutzkommission Band 3266?79, (2007).
[39] Gartel AL, and Tyner AL. 2002. The role of the cyclin-dependent kinase inhibitor p21 in apoptosis. Molecular Cancer Therapeutics 1:639?649, (2002).
[40] Genyao, Y., and Changlin, Y., Multi-organ involvement and failure in a radiation accident: the Chinese experience of 1963. British Journal of Radiology Suppl. 27:55?61, (2005).
[41] George, J.K., Albers, H.E., Carraway, R.E., and Ferris, C.F., Neurotensin levels in the heptic-portal circulation are inversely related to the circadian feeding cycle in

rats. Endocrinology 121:7?13, (1987).
[42] Goans, R.E., Holloway, E.C., Berger, M.E., and Ricks, R.C., Early dose assessment following severe radiation accidents, Health Physics 72:513?18, (1997).
[43] Goodlad, R.A., Al-Mukhtar, M.Y., Ghatei, M.A., Bloom, S.R., and Wright, N.A., Cell proliferation, plasma enteroglucagon and plasma gastrin levels in starved and

refed rats. Virchows Archiv B Cell Pathology Including Molecular Pathology 43:55?62, (1983).
[44] Gorin, N.C., Fliedner, T.M., Gourmelon, P., Ganser, A., Meineke, V., Sirohi, B., Powles, R., and J. Apperley, J., Consensus conference on European preparedness

for haematological and other medical management of mass radiation accidents. Annals of Hematology 85:671?679, (2006).
[45] Grace, M.B., McLeland, C.B., and Blakely, W.F., Real-time quantitative RT-PCR assay of GADD45 gene expression changes as a biomarker for radiation iodosimetry. International Journal of Radiation Biology 78:1011?1021, (2002).
[46] Grace MB, Salter CA, Bullard JR, Prasanna PGS, Manglapus GL, and Blakely WF (2005) Gene-expression biomarkers for application to high-throughput radiation

biodosimetry. Published in the proceedings of the NATO Human Factors and Medicine (HFM) Panel Research Task Group (RTG) 099 Meeting, "Radiation Bioeffects and Countermeasures," Bethesda, MD, USA, June 21?23, 2005. Compact disc: AFRRI CD 05-2.
[47] Grace, M.B., and Blakely, W.F., Transcription of five P53-and Stat-3-inducible genes after ionizing radiation. Radiation Measurements 42:1147?1151, (2007).
[48] Guipaud, O., Holler, V., Buard, V., Tarlet, G., Royer, N., Vinh, J., and Benderitter, M., Time-course analysis of mouse serum proteome changes following exposure

of the skin to ionizing radiation. Proteomics 7:3992?4002, (2007).
[49] Guipaud, O., and Benderitter, M., Protein biomarkers for radiation exposure: towards a proteomic approach as a new investigation tool. Annali dell’Instituto

Superiore di Sanita 45:278?86, (2009).
[50] Guskova, A.K., Baranov, A.E., and Gusev, I.A., Acute radiation sickness: underlying principles and assessment. In: A.E. Gusev, A.K. Guskova, F.A. Mettler, Jr.

(Eds), Medical Management of Radiation Accidents, CRC Press, Washington, DC, pp.33?51, (2009).
[51] Hall, E.J. and Giaccia, A.J., Radiobiology for the Radiologist, 6th ed., Lippincott Williams & Wilkins, Philadelphia, (2006).
[52] Henriksson, R., Frojd, O., Gustafsson, H., Johansson, S., Yi-Qing, C., Franzen, L., and Bjermer, L., Increase in mast cells and hyaluronic acid correlates to

radiation-induced damage and loss of serous acinar cells in salivary glands: the parotid and submandibular glands differ in radiation sensitivity. British Journal of Cancer 69:320?6, (1994).
[53] Hennequin, C., Cosset, J.M., Cailleux, P.E., Girinsky, T., Ganem, G., Hubert, D., Comoy, E., and Dutreix, J., [Blood amylase: a biological marker in irradiation accidents? Preliminary results obtained at the Gustave-Roussy Institut (GRI) and a literature review]. Bulletin du Cancer 76:617?24, (1989).
[54] Hofmann, R., Schreiber, G.A., Willich, N., Westhaus, R., and Bogi, K.W., Increased serum amylase as a probable bioindicator for radiation exposure. Strahlentherapie und Onkologie 166:688?95, (1990).
[55] Huchet, A., Belkacemi, Y., Frick, J., Prat, M., Muresan-Kloos, I., Altan D., Chapel, A., Gorin, N.C., Gourmelon, P. and Bertho, J.M. Plasma Flt-3 ligand concentration correlated with radiation induced bone marrow damage during local fractionated radiotherapy. International Journal of Radiation Oncology Biology and Physics 57:508?515, (2003).
[56] Kashima, H.K., Kirkham, W.R., and Andrews, J.R., Post-irradiation sialadenitis: A study of clinical features, histopathologic changes and serum enzyme variations following irradiation of human salivary glands, American Journal of Roentgenology 94:271?291, (1965).
[57] Katouzian, F., Sblattero, D., Not, T., Tommasini, A., Giusto, E., Meiacca, D., Stebel, M. Marzari, R., Fasano, A., and Ventura, A., Dual sugar gut-permeability testing on blood drop in animal models. Clinica Chimica ACTA 352:191?7, (2005).
[58] Koc, M., Taysi, S., Sezen, O., and Bakan, N., Levels of some acute-phase proteins in the serum of patients with cancer during radiotherapy. Biology Pharmaceutical Bulletin 26(10):1494?1497, (2003).
[59] Koenig, K.L., Goans, R.E., Hatchett, R.J., Mettler Jr., F.A., Schumacher, T.A., Noji, E.K., Jarrett, D.G., 2005. Medical treatment of radiological casualties: current concepts. Annals of Emergency Medicine 45,643?652, (2005).
[60] Leslie, M.D., and Dische, S., Changes in serum and salivary amylase during radiotherapy for head and neck cancer: a comparison of conventional fractionated radiotherapy with CHART, Radiotherapy and Oncology 24:27?31, (1992).
[61] Lichtenberger, L., Welsh, J.D., and Johnson, L.R., Relationship between the changes in gastrin levels and intestinal properties in the starved rat. American Journal of Digestive Disease 21:33?8, (1976).
[62] Liu, W., Ding, I., Chen, K, Olschowka, J., Xu, J., Hu, D., Morrow, G.R., and Okunieff, P., Interleukin 1β (IL1B) signaling is a critical component of radiation-induced skin fibrosis. Radiation Research 165:181?91, (2006).
[63] Lutgens, L., and Lambin, P., Biomarkers for radiation-induced small bowel epithelial damage: an emerging role for plasma citrulline. World Journal of Gastroenterology 13:3033?3042, (2007).
[64] Lutgens, L.C.H.W., Deutz, N.E.P., Gueulette, J., Cleutjens, J.P.M., Berger, M.P.F., Wouters, B.G., von Meyenfeldt, M.F., and Lambin, P., Citrulline: a physiologic marker enabling quantitation and monitoring of epithelial radiation-induced small bowel damage. International Journal of Radiation
Oncology Biology and Physics 57:1067?74, (2003).
[65] Lutgens, L.C.H.W., Deutz, N., Granzier-Peeters, M., Beets-Tan, R., De Ruysscher, D., Gueulette, J., Cleutjens, J., Berger, M.,Wouters, B.,von Meyenfeldt, M., and Lambin, P., Plasma citrulline concentration: a surrogate end point for radiation-induced mucosal atrophy of the small bowel. A feasibility study. International Journal of Radiation Oncology Biology and Physics 60:275?85, (2004).
[66] MacVittie, T.J., Farese, A.M., Jackson 3rd., W., Defining the full therapeutic potential of recombinant growth factors in the post radiation-accident environment: the effect of supportive care plus adminis-tration of G-CSF. Health Phys. 89:546?55, (2005).
[67] Maier, H., and Bibl, H., Effect of radioactive iodine therapy on parotid gland function. Acta Oto-laryngolica 103:318?24, (1987).
[68] Mal’tsev, V.N., Strel’nikov, V.A., and Ivanov, A.A., C-reactive protein in the blood serum as an indicator of the severity of radiation lesion. Doklady Akademii Nauk SSSR 239:750?2, (1978).
[69] Mal’tsev, V.N., Ivanov, A.A. Mikhailov, V.F., and Mazurik, V.K., The individual prognosis of the gravity of the outcome of radiation disease on immunological indexes. Radiatsionnaia Biologiia Radioecologiia [Radiation Biology Radioecology], 46:152?8, (2006).
[70] Marchetti, F., Coleman, M.A., Jones, I.M., and Wyrobek, A.J., Candidate protein biodosimeters of human exposure to ionizing radiation. International Journal of Radiation Biology 82:605?39, (2006).
[71] Martin, M., Vzenin, M.-C., Gault, N., Crechet, F., Pfarr, C.M., and Lefaix, J.-L., Coactivation of AP-1 activity and TGF-β1 gene expression in the stress response of normal skin cells to ionizing radiation. Oncogene 15:981?9, (1997).
[72] Meineke, V., The role of damage to the cutaneous system in radiation-induced multi-organ failure. British Journal of Radiology Suppl. 27:85?99, (2005).
[73] Monti, P., Wysocki, J., van der Meeren, and A., Griffiths, N.M., The contribution of radiation-induced injury to the gastrointestinal tract in the development of multi-organ dysfunction syndrome or failure. British Journal of Radiology Suppl. 27:89?94, (2005).
[74] Muller, K., and Meineke, V., Radiation-induced alterations in cytokine production by skin cells. Experimental Hematology 35:96?104, (2007).
[75] Muller, K, Kohn, F.-M., Port, M., Abend, M., Ring, J., and Meineke, V., Intercellular adhesion molecule-1: a consistent inflammatory marker of the cutaneous radiation reaction both in vitro and in vivo. British Journal of Dermatology 155:670?9, (2006).
[76] Nagler, R., Marmary, Y., Fox, P.C., Baum, B.J., Har-El, R., and Chevion, M., Irradiation-induced damage to the salivary glands: the role of redox-active iron and copper. Radiation Research 147:468?76, (1997).
[77] Okunieff, P., Chen, Y., Maguire, D.J., and Huser, A.K., Molecular markers of radiation-related normal tissue toxicity. Cancer Metastasis Reviews 27:363?74, (2008).
[78] Ossetrova, N.I., Farese, A. M., MacVittie, T.J., Manglapus, G.L., and Blakely, W.F., The use of discriminant analysis for evaluation of early-response multiple biomarkers of radiation exposure using non-human primate 6-Gy whole-body radiation model. Radiation Measurements 42:1158?1163, (2007).
[79] Ossetrova, N.I., and Blakely, W.F., Multiple blood-proteins approach for early-response exposure assessment using an in vivo murine radiation model. International Journal of Radiation Biology 85(10):837?850, (2009).
[80] Ossetrova, N.I., Sandgren, D.J., Gallego, S., Blakely, W.F., Combined approach of hematological biomarkers and plasma protein SAA for improvement of radiation dose assessment in triage biodosimetry applications. Health Physics 98:204?8 (2010).
[81] Petrov, R.V. Immunologiya Ostrogo Luchevogo Porazheniya [Immunology of Acute Radiation Injury]. Gosatomizdat, Moscow, pp. 1?267 (1962); English translation accessed by Joint Publications Research Service, Arlington VA (1963).
[82] Peter, B., Van Waarde, M.A., Vissink, A., 's-Gravenmade, E.J., and Konings, A.W., Radiation-induced cell proliferation in the parotid and submandibular glands of the rat. Radiation Research 140(2):257?65, (1994).
[83] Peter, R.U., Steinert, M., and Gottleber, P., The cutaneous radiation syndrome: diagnosis and treatment. Radioprotection 36(4):451?7, (2001).
[84] Pia de la Maza, M, Gotteland, M, Ramirez, C., Araya, M., Yudin, T., Bunout, D., and Hirsch, S., Acute nutritional and intestinal changes after pelvic radiation. Journal of the American College of Nutrition 20:637?42, (2001).
[85] Roy, L., Berthro, J.M., Squidi, M, Vozenin, M.C., Voisin, P., and Benderitter, M., Biochemical approach to prediction of multiple organ dysfunction syndrome. British Journal Radiology Supplement 27:146?51, (2005).
[86] Tukachinski, S.E. and V. P. Moiseeva. Cx-reactive protein in radiation injury. Bulletin of Experi-mental Biology and Medicine 52:48?52, (1961).
[87] Ulrich, D., Noah, E.M., von Heimburg, D., and Pallua, N., TIMP-1, MMP-2, MMP-9, and PIIINP as serum markers for skin fibrosis in patients following severe burn trauma. Plastic Reconstructive Surgery 111:1423?31, (2003).
[88] Vigneulle, R.M., Rao, S., Fasano, A., and MacVittie, T.J., Structural and functional alterations of the gastrointestinal tract following radiation-induced injury in the rhesus monkey. Digestive Disease Science 47:1480?96, (2002).
[89] Waselenko, J.K., MacVittie, T.J., Blakely, W.F., Pesik, N., Wiley, A. L. Dickerson, W.E., Tsu, H., Confer, D.L., Coleman, C.N., Seed, T., Lowry, P., Armitage, J. O., and Dainiak, N., Medical management of the acute radiation syndrome: recommendations of the Strategic National Stockpile Radiation Working Group. Annual of Internal Medicine 140:1037?1051, (2004).
[90] Yu, H.C., Tugeja, S., Moon J.I., Kleiner, G.I., Conanan, L., Gaynor, J.J., Kato, T., Levi, D.M., Nishida, S., Selvagi, G., Gandia, C., Weppler, D., Esquenazi, V., Ruiz, P., Miller, J., and Tzakis, A.G., Utilization of dried blood spot citrulline level as a non-invasive method for monitoring graft function following intestinal transplantation. Transplantation 80:1729?33, (2005).


04. 2012年8月11日 15:47:46 : ad1KZuILvM
厨さんのこの作業は何時間かかったのだろうか。
両手うちかな。

何かを見つけるだけで、そこから先へは人任せの基の性質にとらわれて。
楽しかった事故以前の生活と、気を緩めると、中途半端に知りすぎた自分の心に負けて、自分が原因で自分の家族さえ破壊しかねない今の自分。

知っていることから判断すると、私と私の子供たちに輝く未来など予測できない。
1年ぶりに、毎日涙が流れ、独りになると、お気楽な嘘に満ち溢れた、白痴と反社会的人格障害が書き連ねる地元新聞の記事に、現実との乖離に嗚咽する。

昔の友は訪ねてきたが、お互い本心を隠せずながらもしらを切り続ける会話。
もっとはっきり言えよ言いたいことを、君の方が私よりずっと上手で、影響力のある地位にいるだろう。

私は、私の子供たちは何のために生を受けたのか。
少なくともこんな馬鹿なやつらのご機嫌取りのためではない。
私の先祖は奴隷ではない。神とのかかわりを誇りにしていたはずだ。

すべてが面倒になってきたが、まだ私はこの結末を見ていない。私が生きてきた意味も知らない。
私はこの結末だけは自分のこの目で見てみたい。
嘘と欺瞞が大好きなやつらがうまくやり通すのか、地獄に落ちるのか。
嘘とペテンにまみれて、地獄に落ちるがいい。お前らとはきっと、人類史の発生当初から別人種に違いない。



05. 2012年8月11日 16:03:33 : ySlPyZ3U5g
泣いてばかりいないで前へ進もう。
私は馬鹿だから事実を見つけ、事実を語るしか出来ない。
まあそれでいいさ、詐欺師になれば並以上の素質があるとは思うけれど、いまさら人生生き方を変えるなんて、めんどくさくてたまらない。

06. 2012年8月11日 16:17:53 : E0X29ePw02
弁証法か、ははは、東海○○さんよりは詳しいかもね。
矛盾がなければ世界はない、か。なんとかアウフヘーベンできそうだ。
スレの皆さんありがとう。この目立たないところにお礼を書いておきます。
みんな、外的作用との交互作用には還元できない内的根拠に突き動かされて生きていく。
私は私。やつらはやつら。
さてまた役立つことを、知恵を投稿するぞ。

07. 2012年8月16日 22:05:22 : NNLqIP7MXD
そうだ、嘘で勝ち取る未来なんて、本当の未来ではない。

08. SaveChild厨 2012年10月03日 01:31:06 : hfhZnjAARar8w : jDO82o1xf2
Chapter 6
Biodosimetry and Biomarkers for Radiological Emergency Response

D. Wilkinson (CAN), D.C.B. Holt (GBR), K. Rothkamm (GBR), A. Jaworska (NOR), W.F. Blakely (USA), D.L. Stricklin (SWE)
Subsection 2 Contribution Authors
E. Waller (CAN), K. Millage (USA), W.F. Blakely (USA), J.B. Nemhauser (USA), Carl A. Curling (USA), Deena S. Disraelly (USA), Ira H. Levine (USA), David J. Sandgren (USA), James A. Ross (USA), J.S. Nasstrum (USA), G. Sugiyama (USA), S. Homann (USA), B.R. Buddemeier (USA)
Subsection 4 Contributing Authors

ABSTRACT
In 2006 NATO Research Task Group HFM-099/RTG-033 addressed the development of new and improved methods to counter the effects of increased risk scenarios such as nuclear/radiological threats. The group’s objective was to develop the scientific basis for the improved methods to prevent, assess, treat, and manage casualties and long-term health effects associated with ionizing radiation exposure. Radiation Injury Assessment and Biodosimetry Subgroup #2 identified a Program of Work outlining specific tasks recognized by all member states as essential areas of interest. These tasks are to: (a) develop an evaluation tool that will guide the selection, advancement, implementation and validation of complementing biodosimetry assays [CAN, GBR, SWE, USA]; (b) develop and validate lead candidate multi-parameter biodosimetry tools [CAN, CZE, DEU, FRA, GBR, NOR, POL, SWE, USA]; (c) evaluate software applications designed to provide radiation injury assessment, biological dosimetry information and guidance to the first-responder and first- receiver communities [CAN, CZE, DEU, FRA, GBR, SWE, USA]; (d) develop injury assessment and biological dosimetry guides and documents [GBR, USA]; and (e) develop a proposal for Advanced Demonstration Technology by the final RTG-099 meeting [CAN, CZE, DEU, FRA, GBR, NOR, POL, SWE, USA].

1.0 INTRODUCTION
NATO recognizes that current physical and biomedical scientific support is not sufficient to provide the necessary advice for consequence management and medical countermeasures in scenarios involving radio- logical exposures. Research Task Group HFM-099/RTG-033 undertook the development of new and im-proved methods/tools to assist the medical community in responding to radiological/nuclear events involving casualties. The group‘s objective was to develop the scientific basis for new and improved methods to prevent, assess, treat, and manage casualties and long-term health effects associated with ionizing radiation exposure. Five specific tasks were identified by the group as targets for advanced research. Efforts undertaken within each of these tasks are described in this summary document.
2.0 DEVELOP AN EVALUATION TOOL THAT WILL GUIDE THE SELECTION, ADVANCEMENT, IMPLEMENTATION AND VALIDATION OF COMPLEMENTING BIODOSIMETRY ASSAYS [CAN, GBR, SWE, USA, FIN, NOR]
2.1 Biodosimetry Assay Evaluation Tool Medical and scientific communities are well aware of the challenges to be encountered in management of Radiological/Nuclear mass casualty disasters. Available medical resources will need to be stringently controlled and directed towards optimal outcomes. Identification of casualties needing medical intervention would be the primary challenge for the medical community. At the present time, there is no biodosimetry assay that is suitable for all scenarios. The selection of the most appropriate assay or assays will depend on the scenario circum-stances, the number of casualties that need to be assessed, and the available resources. An ideal dosimeter, or more likely a combination of the most suitable dosimeters, may need to be identified for each scenario. The task of this working group is to provide cumulative information on different assays available and rate each one according to the NATO Technology Readiness Level (TRL) Assessment guide.
It is hoped that the outcome of is process will result in a tool that will assist casualty management controllers in their selection of the most appropriate and relevant biodosimetry tools for the particular scenario and will guide the international research programs for improved methods based on the TRL evaluation process.
The initial approach to facilitating this tool development was to first identify all possible assays and then categorize them according to their function properties. The next step was to rate each assay according to the NATO‘s TRL guide (Table 1).

The first step, identification of all possible assays, was relatively easy because the Working Group participants had a broad understanding of existing and surfacing technologies. The list of potential assays was all- encompassing and became too large for a single table format. It was agreed that the assays needed to be sorted according to their functional properties. Four separate functional property groupings were established: 1) prodromata; 2) haematological/biochemical; 3) cytogenetic; and 4) physical. Within each functional area, technologies/assays were characterized by a number of Rating Criteria; these were the same in all functional groups for all technologies (Specificity, Sensitivity, Low Background, Low Donor Variability, Doubling Dose, Dose Response Calibration, Persistent Effect, Ease of Sampling, Early Sampling, Ease of Analysis, Rapid Analysis, Simplicity of Analysis, Low Inter-observer Error Probability, Low Intra-observer Error Probability, Cost, Risk Meter, Relative Biological Effect (RBE), Partial Body Exposure, Automation, and Deployable). Once the criteria were established, the Working Group members evaluated each Rating Criteria using the TRL guide. The average TRL was determined by averaging the TRL scores for all Rating Criteria in a given technology. The confidence of each assay was determined by tabulating the total number of Rating Criteria against the accompanying average TRL. This cumulative evaluation of the available technologies is presented in Figures 1 through 4.
Table 1:
Technology Readiness Level (TRL) Assessment guide

0

Basic Research with Future Military Capability in Mind

1

Basic Principles Observed and Reported in Context of a Military Capability Shortfall

2

Technology Concept and / or Application Formulated

3

Analytical and Experimental Critical Function and / or Characteristic Proof of Concept

4

Component and / or “Breadboard” Validation in a Laboratory / Field (e.g., ocean) Environment

5

Component and / or “Breadboard” Validation in a Relevant (operating) Environment

6
System / Subsystem Model or Prototype Demonstration in a Realistic (operating) Environment or Context

7

System Prototype Demonstration in an Operational Environment or Context (e.g. exercise)

8

Actual System Completed and Qualified through Test and Demonstration

9

Actual System Operationally Proven through Successful Mission Operations The first step, identification of all possible assays, was relatively easy because the Working Group participants had a broad understanding of existing and surfacing technologies. The list of potential assays was all- encompassing and became too large for a single table format. It was agreed that the assays needed to be sorted according to their functional properties. Four separate functional property groupings were established: 1) prodromata; 2) haematological/biochemical; 3) cytogenetic; and 4) physical. Within each functional area, technologies/assays were characterized by a number of Rating Criteria; these were the same in all functional groups for all technologies (Specificity, Sensitivity, Low Background, Low Donor Variability, Doubling Dose, Dose Response Calibration, Persistent Effect, Ease of Sampling, Early Sampling, Ease of Analysis, Rapid Analysis, Simplicity of Analysis, Low Inter-observer Error Probability, Low Inta-observer Error Probability, Cost, Risk Meter, Relative Biological Effect (RBE), Partial Body Exposure, Automation, and Deployable). Once the criteria were established, the Working Group members evaluated each Rating Criteria using the TRL guide. The average TRL was determined by averaging the TRL scores for all Rating Criteria in a given technology. The confidence of each assay was determined by tabulating the total number of Rating Criteria against the accompanying average TRL. This cumulative evaluation of the available technologies is presented in Figures 1 to 4.

Figure 1: Prodromata Functional Area. Each point represents different Technology/Assay listed in the table legend and plotted against the Total Number of Criteria Scored. Rating Criteria evaluations for Prodromata Functional Area included: Specificity, Sensitivity, Low Background, Low Donor Variability, Doubling Dose, Dose Response Calibration, Persistent Effect, Ease of Sampling, Early Sampling, Ease of Analysis, Rapid Analysis, Simplicity of Analysis, Low Inter-observer Error Probability, Low Intra-observer Error Probability, Cost, Risk Meter, Relative Biological Effect (RBE), Partial Body Exposure, Automation, and Deployable. Note.
FRAT: First-responders Radiological Assessment Triage.

Figure 2: Haematological/Biochemical Functional Area. Each point represents different Technology/ Assay listed in the table legend and plotted against the Total Number of Criteria Scored. Rating Criteria evaluations for Haematological / Biochemical Functional Area included: Specificity, Sensitivity, Low Back- ground, Low Donor Variability, Doubling Dose, Dose Response Calibration, Persistent Effect, Ease of Sampling, Early Sampling, Ease of Analysis, Rapid Analysis, Simplicity of Analysis, Low Inter-observer Error Probability, Low Intra-observer Error Probability, Cost, Risk Meter, Relative Biological Effect (RBE), Partial Body Exposure, Automation, and Deployable. Note.
BAT (LDK): Biodosimetry Assessment Tool ? lymphocyte depletion kinetics algorithm.

Figure 3: Cytogenetic Dosimetry Functional Area. Each point represents different Technology/Assay listed in the table legend and plotted against the Total Number of Criteria Scored. Rating Criteria evaluations for Cytogenetic Dosimetry Functional Area included: Specificity, Sensitivity, Low Background, Low Donor Variability, Doubling Dose, Dose Response Calibration, Persistent Effect, Ease of Sampling, Early Sampling, Ease of Analysis, Rapid Analysis, Simplicity of Analysis, Low Inter-observer Error Probability, Low Intra-observer Error Probability, Cost, Risk Meter, Relative Biological Effect (RBE), Partial Body Exposure, Automation, and Deployable. Note. PCC: premature chromosome condensation; RICA: rapid interphase chromosome aberration assay.

Figure 4: Physical Dosimetry Functional Area. Each point represents different Technology/Assay listed in the table legend and plotted against the Total Number of Criteria Scored. Rating Criteria evaluations for Physical Dosimetry Functional Area included: Specificity, Sensitivity, Low Background, Low Donor Variability, Doubling Dose, Dose Response Calibration, Persistent Effect, Ease of Sampling, Early Sampling, Ease of Analysis, Rapid Analysis, Simplicity of Analysis, Low Inter-observer Error Probability, Low Intra-observer Error Probability, Cost, Risk Meter, Relative Biological Effect (RBE), Partial Body Exposure, Automation, and Deployable. Note. TLD: Themoluminescence dosimetry: OSL: optically stimulated luminescence.
2.2 Interpretation of the Results

Each Functional Area (Figures 1 to 4) presented a distribution of TRLs for the different technologies/assays evaluated. For Prodromata (Figure 1), the top scoring assays were: 1) Time of, or to, Vomiting; and 2) Time of, or to, Nausea. Time of, or to, Vomiting had the highest average TRL score (7.7), while Time of, or to, Nausea scored second highest with an average TRL of 6.2. The total criteria evaluated for both of these technol- ogies/assays were the greatest of all Prodromata, indicating the highest evaluator confidence. Most of the other technologies/assays evaluated had a much lower value for total number of criteria scored suggesting a lower confidence in these assays. Therefore, these other assays had an overall lower score even though they may have been scored high on the TRL scale. For Haematological/Biochemical technologies/assays (Figure 2), the top scoring assay was the Lymphocyte Depletion Kinetics with an average TRL score of 7.1 and the highest evaluator confidence.
Other assays may havred slightly higher on the TRL scale but fewer criteria were able to be evaluated indicating lower evaluator confidence. The Dicentric Assay scored the highest with an average TRL of 6.6 in the Cytogenetic Functional Area (Figure 3). The total number of criteria scored for all Cytogenetic Functional Area assays was very similar, and indicated a very high evaluator confidence level for all evaluated technologies/assays.
Figure 4 represented evaluation data for Physical Functional Area. Of all the technologies listed (Film badge, thermoluminescence dosimeter (TLD), optically stimulated luminescence (OSL), Neutron (Track H-polyallyldiglycol carbonate (PADC)), Neutron (Bubble), Neutron (Albedo effect single or multiple element), only three were evaluated. Although the confidence of the evaluations was low, the average TRL for all three physical technologies was very high (between 7.5 and 7.7). Based on the data analysis using the limited evaluator information, it can be concluded that the highest average TRLs were achieved with Physical Dosimetry, followed by Prodromata, Cytogenetic and then Heamatological/Biochemical technologies/assays. However, the highest evaluator confidence was observed in the Cytogenetic Functional Area followed by Haematological/Biochemical, Prodromata and then Physical. It is important to note that the Working Group participants contributing to the evaluation of this tool had an expertise bias in the biological Functional Areas. However, it is also important to note that medical professionals do not treat a dose but rather the symptoms of a dose exposure, and as such physical dosimeters alone should not dictate specific medical treatment decisions.
2.3 Critical Evaluation of the Tool

The developed Tool presented in Figures 1 to 4 and discussed in Section 2.2 is intended for guiding the selection, advancement, implementation and validation of complementary biodosimetry assays. Although this evaluation of cumulative data as a tool has merit and could lead to useful information, significant challenges
need to be overcome before this methodology would become fully useful. Some of these challenges include: 1. Differing Requirements by End-users: Early in the evaluation process, the question arose as to who would be using this information and how to provide this information in the most useful template. Initially it was
thought that this information would be used by the Military Planners and Medical First Responders for the purpose of planning and responding to Radiological/Nuclear emergency events. As a tool targeted to the medical community and planners, it had to be user friendly without a specific requirement for scientific details.

In addition to using this tool during emergency response scenarios and for planning, the question was also posed whether the provided information may be useful in the long-term medical management of irradiated casualties. Early and ongoing guidance on appropriate selection of assay methodologies may be required in a mass casualty disaster. This requirement may vary depending on the number of casualties, the type of injuries and the available resources. Again, in this case there would be no need for detailed scientific information.

The final application for this tool was to develop a TRL-based survey of presently available and upcoming technologies that would guide the directed funding and future research of the scientific community. For this requirement, a more detailed scientific overview was required 2. Coverage of Functional Areas: All presently available or upcoming technologies/assays were grouped according to four Functional Areas: 1) Prodromata; 2) Haematological/Biochemical; 3) Cytogenetic; and 4) Physical. The question was raised as to the completeness in coverage by these four functional areas and if there is a need for adding the Biophysical Functional Area (i.e. Radiobioassays). Biophysical Functional Area would deal with body fluid samples such as urinary, faecal, nose swabs and blows, and wound swabs. Due to the complexity of analysis, high dependence on the physico-chemical properties of the contaminating radionuclides, and the route of entry, this additional information could be meaning-less without expert interpretation. Even though it was decided to abstain from including this massive, unmanageable information, it was deemed an important Functional Area that needed to be recognised.
3. Assignment of TRL values: As the process evolved, it became very evident that it was difficult to assign a TRL value to a number of Rating Criteria characterizing each technology/assay. The TRL was calcu-lated by determining the average TRL across all criteria rated. Some responders felt that it was too dif-ficult to assign a TRL-based value to many of the Rating Criteria. Others suggested that the challenge
of assigning the most appropriate TRL to each criterion would require significant analysis by an extensive team of experts, and as a consequence they chose to abstain from the evaluation process.

Additionally, a question was posed: Does each of the evaluated Rating Criteria have equal weight or are some criteria more important than others? This biased weighting would result in different TRL values than those presented in Figures 1 to 4 and discussed in Section 2.2. The assignment of appropriate weight-ing factors to each of the Rating Criteria is foreseen as a daunting task with a potential for significant debates.
4. Selection of Rating Criteria: Aside from assigning the TRL value to each criterion, there was also some concern with the selected Rating Criteria. The Rating Criteria for each technology/assay in each Functional Area are the same. They are: Specificity, Sensitivity, Low Background, Low Donor Variability, Doubling Dose, Dose Response Calibration, Persistent Effect, Ease of Sampling, Early
Sampling, Ease of Analysis, Rapid Analysis, Simplicity of Analysis, Low Inter-observer Error Probability, Low Intra-observer Error Probability, Cost, Risk Meter, Relative Biological Effect (RBE),
Partial Body Exposure, Automation, and Deployable. Some of the reviewers questioned the appropriate-ness of applying the same Rating Criteria against some technologies/assays. In some cases not all Rating
Criteria were given a TRL. For this reason the total number of criteria rated by all evaluators was different for different technologies/assays.
5. Lack of Evaluator Response: Early in the process it became evident that despite the diligent efforts of the participants, there was a strong hesitation to complete the evaluation process. The lack of sufficient
responses indicated that the resulting evaluation process may be biased and in line with opinions of very few responders; in all cases only 3 or fewer data sets contributed to the evaluation of average
TRLs. This would suggest that the presented TRLs in Figures 1 to 4 may not be representative of a broader scientific community.
6. Bias in Expertise: There was a strong bias in evaluating the rating criteria for the biological techno- logies/assays (Figures 1 to 3), as discussed in Section 2.2. Comparatively, expertise in evaluating the

Physical Functional Area technologies was lacking.
TRL Variation: Finally, even with a small number of respondents, it was evident that the experts contributing to the evaluation process had different opinions on the appropriate TRL for many of the Rating Criteria. In
some cases there was a great variance in the criteria scored and the allocated TRLs.
Proposed Solutions/Recommendations

1. The proposed tool has a strong potential for providing ―finger tip? information to medical profession- als, emergency planners and the supporting scientific community. For maximum impact, the tool
would need to be expanded to provide more detailed information for the scientific community and the funding agencies, and then compressed for rapid and easy access by the medical professionals and
emergency planners. In both circumstances, for the tool to be of value, it would have to be kept as current as possible with the latest scientific developments.
2. It is recognized that the tool is lacking the Biophysical Functional Area. The information gained through Biophysical technologies/assays is of significant importance and needs to be acknowledged.
Useful integration of this information can only be achieved through the use of computer automation.
3. The process of developing this tool was based on evaluating the same Rating Criteria across different Functional Areas. One recommendation may be to introduce unique and most appropriate Rating Criteria
for each Functional Area. Moreover, even though it may be justifiable to introduce calculated weighting factors for different Rating Criteria, it is very clear that such activity would be subject to broad debate
and scrutiny and therefore not feasible.
4. Finally, for this assay to be most useful, the data input would need to come from a much broader group of experts, including experts in physical and biophysical dosimetry. By expanding the number of
evaluators who bring different expertise it is likely that the calculation of average TRL values would be more representative of the broader scientific community. Increasing the number of evaluators
may result in a more realistic and non-biased calculations of TRLs and also provide some prediction of opinion-deviances.
2.4 Summary
The proposed tool intended to guide the selection, advancement, implementation and validation of complementing biodosimetry assays is most likely to be of greatest benefit to the community of biodosimetry
experts who may in turn support and advise medical and emergency response professionals. The information gathered through contributions from an extensive group of experts could lead to a very useful tool that would
need to be continually updated and maintained to provide the best information. Finally, it is important to remember that the final selection of appropriate technologies/assays will depend on
the unique scenario, the available resources and expertise, and on the interactive communications among many disciplines.
3.0 DEVELOP AND VALIDATE LEAD CANDIDATE MULTI-PARAMETER BIODOSIMETRY TOOLS [CAN, CZE, DEU, FRA, GBR, NOR, POL, SWE, USA] Medical management of radiation casualties necessitates the use of multiple parameter biological dosimetry
assessment. Even more desirable (and more difficult to accomplish) is the identification of parameters (biomarkers) predicting the extent of radiation induced cell damage, the biological response to it and finally
the prognosis.

Figure 5: Summary on RTG033 efforts for elucidating gene expression significance for biodosimetry. In vitro models either comprise irradiated peripheral mononucleated cells (PMNC) or peripheral blood lymphocytes (PBL). No single parameter is sufficient to cover the large variety of possible scenarios (from dirty bomb to nuclear
weapon) and specific requirements on the measurements. For acute radiation effects, results within the first 4 days after exposure are required. In case of late effects, measurements performed years after exposure should
provide some clues to the potential exposure. The dicentric chromosomal aberration assay (DCA assay) represents the most established assay for dose estimates. As an effort of RTG033 this kind of analysis was combined with known dose dependency of acute
clinical signs/symptoms occurring after radiation exposure together with physical dosimetric measurements and other parameter to a software called BAT (Biodosimetry Assessment Tool). Still, this approach
necessitates inputs from further areas in order to combine the strength of different assays and to compensate for their limitations.
Nowadays, there is overall agreement on the potential of radiation associated gene expression changes for biodosimetry (Amundson 2000, 2001, Blakely 2003, Grace 2002). Less is known on the prediction of
radiation induced cell damage by radiation associated gene expression changes. Candidates like GADD45A, CDKN1A (p21) or Bax showed a dose-response relationship in a large variety of different in vitro and in vivo
models. Besides these promising findings a systematic approach is needed for contributing to the task described above. This includes a meaningful combination of in vitro and in vivo models together with a corresponding sequence of different gene expression methods and platforms (Figure 5).
Different nations (FRA, GER, USA) exchanged results on gene expression within the framework of RTG033 and plan to continue working together on future joint projects. As a result of these efforts a variety of in vitro
and in vivo models using different exposures and platforms for gene expression analysis evolved (Figure 6). The concept, interplay and results (summarized) of these efforts are shown below.

Figure 6: Fold-changes in radiation associated GADD45A gene expression relative to control utilizing different in vitro models and time points up to 48 h after irradiation. PMNCs were incubated
at different conditions (room temperature, left graph or at 37°C remainder two graphs) and GADD45A gene expression was measured either on PMNCs or at CD4+ lymphocytes only, as indicated in the
graphs. Data in the left and right graphs represent unpublished data (GER). Fold-changes of PMNC incubated at 37°C (graph in the middle) are mean values examined in three individuals and are drawn
from RTG033 2005 report (Grace et al.). Symbols represent mean values and error bars are SEM (n=30 for PMNC at room temperature and n=6 for CD4+ lymphocytes incubated a 37°C).
3.1 Exposures
Exposures using different quality of radiation, doses and desirably different dose rates are needed in order to cover a large variety of possible exposure scenarios. AFRRI provided and will continue to provide the
professional support (physical dosimetry department) requirements. A USA study on primates irradiated with sub-lethal and supra-lethal doses was completed in 2009 with samples archived for GER to perform a 2-stage
study design. First, a whole genome screening utilizing a microarray will be done to search for radiation associated up- and down- regulated genes. Second, the ―h ot candidate genes? resulting from the microarray
screening will be examined quantitatively using a certain high throughput quantitative real-time PCR platform (TaqArray or low Density Array, LDA).
3.2 In Vitro/In Vivo Models
Three in vitro models were established (Figure 5). These models probably depict different aspects of in vivo models; however these in vitro models need to be validated against in vivo models. In this context the
following features must be considered. Radiosensitivity of blood subpopulations differs according to the following sequence (from highest to lowest sensitivity): B-lymphocytes > CD4+
lymphocytes > CD8+ lymphocytes > NK cells (Louagie et al. 1999).
The number within each blood subpopulation differs among individuals.
The blood in the human body is circulating. This facilitates an interaction with other body parts to occur.
The peripheral blood represents only one compartment of the hematopoietic system. Examinations focusing on peripheral blood do not cover other compartments such as the bone marrow or lymphatic
system. About 1% of all lymphocytes are circulating in the periphery and about 50% are located in the gastrointestinal tract. Radiosensitivity of the hematopoetic system is higher than for most other organ systems.

In other words, the significance of examinations of whole blood in vitro models is limited. Only by compari-son with results from in vivo models can their impact for biodosimetry be judged.
In a first attempt, all contributing nations examined whether a dose response relationship of already assumed/known radiation-induced genes using semiquantitive microarrays (Amundson et al. 2000) can be
demonstrated/shown in their in vitro models, by utilizing more quantitative methods (RTQ-PCR). In particular GADD45A (and others) proved to be of significance (Figure 6 and Grace et al. RTG033 2005 report).
Nevertheless, the pros/cons of the in vitro models (Table 2) necessitate in vivo examinations using in vivo models. Provided the agreement between the models is satisfying, further examinations (e.g. changes in dose
rate) could continue to be performed in in vitro models. This strategy amplifies the task of RTG033 by significantly reducing the number of animal experiments as much as possible. Hence, cooperation between partners combining their expertise will enable joint experiments, as outlined above, in the forthcoming USA primate study. An ongoing French-German cooperation examines gene expression changes on irradiated swine epidermis.

Table 2: Characteristics (pros/cons) of different in vitro models using peripheral mononucleated cells (PMNC) or isolated peripheral blood lymphocytes (PBL with CD4+/CD8+).
3.3 Platforms/Chemistries
A large variety of commercially available chemistries and platforms can be utilized for gene expression-based biodosimetry. The endpoint for these models is gene expression changes measured in peripheral blood after radiation exposure. RNA is a sensitive molecule and can degrade easily because of ubiquitously existing RNases (e.g., contamination from hands).
PAXGene refers to a system which lyses the peripheral blood after venipuncture using Bectin Dickerson‘s Vacutainer R technology (Bectin Dickerson, Franklin Lakes, NJ, USA).
Using this technology platsform RNases become inhibited thus stabilizing the RNA and gene expression changes induced in vivo become ― frozen?. However, cell subpopulations ( e.g., lymphocytes) can not be selected with this method. This method provides an overall picture for gene expression changes occurring within the whole peripheral blood. For selection of cell subpopulations CPT-tubes could be utilized. After venipuncture using a CPT Vacutainer
R system these tubes are centrifuged leading to a physical separation of serum and PMNC from erythrocytes.
This system is stable for 2 days at room temperature. Since it is not required to open the tubes for this procedure the samples remain sterile. Interestingly, cells remaining in these tubes provide an opportunity for examining them under almost normal conditions, such as human serum, without adding artificial substitutes.
Artificial substitutes (e.g. fetal calf serum, RPMI 1640 medium, antibiotics) are added into plastic dishes for in vitro culturing when collecting peripheral blood in EDTA tubes. Although this approach sounds very
artificial it represents the most common procedure, presumably due to decades of experience with this system for the purpose of examining another endpoint (i.e. the dicentric chromosomal aberrations). This approach
was slightly changed in a model established by the German contributor by adding 10% of human serum to the cell culture.
For quantification of gene expression changes occurring in already known radiation induced genes, a single or a multiplex quantitative RT-PCR is utilized (USA, GER). The latter would be preferable, because of
lower costs and probably higher precision (Grace et al, 2003), but additional experiments for establishing a quadruplex RTQ-PCR (USA), for example, are required.
For quantification of gene expression changes occurring in unknown radiation induced genes, a two-stage study design was developed (USA, GER). A genome wide screening with microarrays allows for detection of
potential candidate genes. In the second stage, the gene expression of these genes becomes quantified using a high-throughput RTQ-PCR platform called TaqArray. This platform allows quantifying gene expression of
384 genes at the same time. Details are described in the final report SG1.
This two stage study was utilized in one in vitro model (GER) and is planned to be expanded into the forthcoming primate model. Data are currently under examination. Interesting, about 50% of the candidate
genes selected reveal no changes in gene expression levels.
3.4 Summary
In summary, the significance of already established in vitro models has to be validated by complementary in vivo experiments in near future. This next step will allow us to go back to the validated in vitro models in order to
continue more detailed examinations on the dose response relationships, but avoiding further animal models.
4.0 EVALUATE SOFTWARE APPLICATIONS DESIGNED TO PROVIDE RADIATION INJURY ASSESSMENT, BIOLOGICAL DOSIMETRY INFORMATION AND GUIDANCE TO THE FIRST-RESPONDER AND FIRST-
RECEIVER COMMUNITIES [CAN, CZE, DEU, FRA, GBR, SWE, USA]
There are numerous software tools available for field deployment, reach-back, training and planning use in the event of a radiological or nuclear (RN) terrorist event. Specialized software tools used by CBRNe responders can increase information available and the speed and accuracy of the response, thereby ensuring that radiation
doses to responders, receivers, and the general public are kept as low as reasonably achievable. Software designed to provide health care providers with assistance in selecting appropriate countermeasures or
therapeutic interventions in a timely fashion can improve the potential for positive patient outcome. Several software packages are described in this section and by Waller et al., 2009; although the list of software
presented here is not exhaustive, it does provide a reasonable overview of the types of materials available to the NATO community.
Software tools can be categorized in different manners. One method is to categorize by end user such as: first- on-scene, CBRNe responder, incident commander, health physics reach-back, hospital emergency services,
biodosimetrist, or forensics criminal investigator. A second method is by application use such as: hazard prediction, human effects estimation, and medical triage, dosimetry and treatment. Regardless of the
categorization method used, some of the software tools will fit into multiple categories. It should be noted that since these software tools are, in general, developed for a particular target user, it is very important that they
be used within their intended scope and that users be appropriately trained.
4.1 Hazard Prediction Models
The first group of software tools described in this section are fundamentally hazard prediction models. The codes will allow the user to define an incident or source term and using various transport and dispersion
models, will predict the resultant plume or hazard area. The models estimate the resultant dose or dose rates that might be expected as a result of the dispersed material and some of the models will also
estimate the human response to the dose. Many of these models will perform similar calculations for chemical and biological hazards.
4.1.1 Hazard Prediction and Assessment Capability (HPAC)
The Hazard Prediction and Assessment Capability (HPAC) is a software application that models the transport and dispersion of chemical, biological, radiological and nuclear (CBRN) releases into the atmosphere and
predicts the effects of those hazards on civilian and military populations. HPAC was first released in 1992 and continues to be improved through development funding by the Defense Threat Reduction Agency (DTRA).
HPAC includes several integrated source terms for chemical and biological hazards, as well as for radiological dispersal devices (RDDs), nuclear facility accidents, and nuclear weapon detonations. The transport engine for
the HPAC software, a second-order closure, integrated puff (SCIPUFF) model, is based on a 3-dimensional puff methodology. The transport and dispersion calculation utilizes local terrain effects, including urban
terrain, and can utilize real-time weather input. The airborne concentration and downwind deposition of the hazard plumes are calculated and the results can be used to predict human effects. As a result, the model can
plot not only airborne and deposited activity concentrations, but also internal and external dose estimates. In addition, casualty predictions are estimated based on both prompt nuclear weapon effects and protracted
radiation exposure.
HPAC is used by both military and civilian users for planning, training and exercises, as well as for real-time assessments of on-going incidents. Requests for HPAC software can be made at the following website:
https://acecenter.cnttr.dtra.mil/registration/mainpage.cfm.
4.1.2 HotSpot


09. 2012年10月03日 01:37:08 : jDO82o1xf2
The HotSpot codes provide emergency response personnel and emergency planners with a fast, field-portable set of software tools for evaluating radioactive airborne hazards using a Gaussian plume model. The software is also used for safety analysis of facilities handling nuclear material. Hotspot provides a fast and usually conservative means for estimation of the radiation effects associated with the short-term (less than 24 hours) atmospheric release of radioactive materials over short distances (less than 10 kilometers). HotSpot can be used for quick, initial estimates of the near-term, near-field dose and effects.
The HotSpot plume model has options for a general point source, fire dispersal and explosive dispersal of radioactive material. Dose can be calculated due to plume passage, inhalation, submersion, ground shine and
resuspension, as well as ground deposition. A nuclear explosion model calculates fallout dose and dose rate, and prompt nuclear effects, blast overpressure, thermal radiation, and ionizing radiation. HotSpot incorporates
Federal Guidance Reports 11, 12, and 13 (FGR-11, FGR-12, FGR-13) Dose Conversion Factors (DCFs) for inhalation, submersion, and ground shine. In addition to the inhalation 50-year Committed Effective Dose
Equivalent DCFs, acute (1, 4, 30 days) DCFs are available for estimating deterministic effects. This acute mode can be used for estimating the immediate radiological impact associated with high acute radiation doses
(applicable target organs are the lung, small intestine wall, red bone marrow, and thyroid).
The Hotspot program has an exercise mode that can help illustrate what the responder‘s instrument would actually be reading at the scene of an incident. Maps can be imported into HotSpot, and the contamination
contours can be overlaid on them. Virtual instrument readings can be provided by attaching a GPS unit to the computer or by simply hovering the cursor over the point of interest on the map. Many common instruments
are programmed into HotSpot, and the user has the ability to add instruments used by their response team.
Special purpose programs are also included in HotSpot, including FIDLER Calibration and Lung Screening, and Radionuclides in the Workplace.
HotSpot is maintained by the National Atmospheric Release Advisory Center (NARAC) at Lawrence
Livermore National Laboratory (LLNL) for the U.S. Department of Energy National Nuclear Security
Administration‘s Office of Emergency Response. The HotSpot software is available via Web download:
https://www-gs.llnl.gov/hotspot.
4.2 Human Effects Estimation Models The next group of codes can be categorized as human effects estimation models. They provide guidance concerning the human effects from radiation environments, and in some cases, include blast and thermal environments from nuclear weapon detonations as well. The models provide estimates of the types and levels of injuries that might be expected and some predict the expected patient streams and medical resource require-ments. The codes are generally used for planning purposes, but some could be used during an actual event.
4.2.1 Combined Human Response Nuclear Effects Model (CHRNEM)
The Combined Human Response Nuclear Effects Model (CHRNEM) is a DOS software application that models combined injury from nuclear weapon effects, estimating mi l i t ary combat ant response to simultaneous ionizing radiation (R), blast overpressure (B), and thermal flash burn (T) insults. The effects of these injuries are expressed in terms of performance over time?a soldier‘s continued ability to perform physically undemanding and demanding tasks following a nuclear event. After applying a series of insult and performance-based rules, the user can estimate the total number of people who are injured and become casualties, the number of casualties caused by types of insult (i.e. R-B-T or R-T casualties), and the number of fatalities over time. (Levin 1993, Levin and Fulton 1993)
The program uses a three-dimensional logarithmic interpolation routine for performance estimation at various insult levels. The program includes related algorithms for evaluating symptoms vs. dose and time, crew performance, equivalent prompt dose from a detonation plus fallout event sequence, mortality probability, and early transient incapacitation probability. Psychological or ―battle fatigue? casualties are estimated with an independent technique based on expert evaluation and data analysis for highest correlation to actual battle experience.
CHRNEM includes estimates of nuclear environments as a function of range from a low air burst generic fission weapon of yields ranging from 0.1 to 500 kT, or the user can put in their own environment estimates.
Any values of radiation, thermal, and blast (within the permitted ranges) can be entered instead of using the weapon model. Multiple values of one of these can be accommodated to produce families of performance vs.
time curves when plotted. Both plotted and tabular results can be produced. CHRNEM will also estimate psychological casualties (battle fatigue cases) based on an algorithm which considers the population density and cohesiveness, whether or not there is a warning of the nuclear event, and the yield of the weapon.
CHRNEM has not been accredited by the DoD, although it has been in use for several years both within the United States and other countries. Although questions on the validity and quality of the tool have arisen, CHRNEM and related models may be the best available representations for conducting combined injury estimates following a nuclear detonation.
4.2.2 Radiation Induced Performance Decrement (RIPD)
The Radiation Induced Performance Decrement (RIPD) software is an application that models the probability of injury and mortality from protracted exposure to a radiation field. The RIPD Lethality and Injury Probability Interpolation (RIPDLIPI) is a fast-running tool based on RIPD calculations for the radiation dose rate profile associated with exposure to fallout from a nuclear etonation. RIPDLIPI is the casualty estimation tool for protracted radiation exposure implemented into the Hazard Prediction and Assessment Capability (HPAC) software package. The RIPD code uses a quantitative description of the sign/symptom severities of acute radiation sickness (ARS) developed in the early ?80s. The six categories of the major signs and symptoms of ARS are: 1) Upper Gastrointestinal Distress (UG), 2) Lower Gastrointestinal Distress (LG), 3)
Fatigability and Weakness (FW), 4) Fluid Loss and Electrolyte Imbalance (FL), 5) Infection and Bleeding (IB), and 6) Hypotension (HY). The RIPD code estimates a probability of injury based on the incidence of UG and FW symptoms. It calculates a severity level for each of the six sign/symptom categories, which are in turn used to estimate decrements in human performance capability. The RIPD code also estimates the incidence of mortality and the typical time of mortality using a bone marrow cell kinetic model. The target users of RIPD are both military and civilian, and RIPD is most useful for planning, training and exercises, although it may be used for real-time assessments of on-going incidents.
4.2.3 Nuclear, Biological, and Chemical Casualty Resource Estimation Support Tool (NBC CREST)
The medical Nuclear, Biological, and Chemical Casualty Resource Estimation Support Tool (NBC CREST) performs two key functions: casualty estimation for an NBC scenario and analysis of resource allocation necessary to effectively treat those casualties. The casualty estimation capability will not only predict the number of injuries and fatalities, but also the level of the injury, and the time-dependent nature of the injury progression. The casualty calculations include the individual and combined effects from the blast overpressure, thermal pulse and prompt radiation from a nuclear weapon event. The resource estimation module uses information from the Defense Medical Standardization Board (DMSB) Task, Time and Treater (TTT) files and Deployable Medical Systems (DEPMEDS) databases to compute the time-phased resource requirements for the casualty stream. These data are presented as a series of Excel spreadsheets that can be manipulated by a logistician. The types of resources estimated include bed usage, equipment, blood and other materiel, personnel by occupational specialty, transports, and evacuation resources. The tool can assign the resource consumption to specific medical treatment facilities, allowing the medical planner to optimize the medical footprint and the evacuation network by performing an iterative analysis to eliminate resource
shortfalls. NBC CREST was developed in conjunction with the Army Office of the Surgeon General and the Defense Threat Reduction Agency (DTRA). Currently, NBC CREST is primarily targeted for military users for planning, training and exercises, as well as for real-time assessments of on-going incidents.
4.3 Tools for Medical Recording, Dose Estimation and Treatment Guidance
The next few codes can be used for real-time medical recording (i.e., BAT), dose-estimation (i.e., BAT, FRAT) and treatment guidance (i.e., MEDECOR) during an event. The software tools provide a means of recording relevant clinical data (lymphocyte counts), as well as prodromal signs and symptoms (time to emesis) and will predict the likely dose levels that the patient may have been exposed to. The tools are not meant to direct medical treatment, but some will provide suggestions regarding treatment options and decorporation techniques.
4.3.1 Biodosimetry Assessment Tool (BAT)
The Biodosimetry Assessment Tool (BAT) is a comprehensive software application developed by the Armed Forces Radiobiology Research Institute (AFRRI) for recording patient-specific diagnostic medical information in suspected radiological exposures (Sine et al., 2001; Salter et al., 2004). An updated application (Version 1.03) was released in September 2007. BAT operates under the Microsoft Windows operating system and provides data-collection templates for dynamically recording data in seven folders including: physical dosimetry, contamination/wound, prodromal symptoms, hematology, lymphocyte cytogenetics, erythema, and infection. The software application includes the additional features of dose assessment algorithms based on:
i) onset of vomiting, ii) lymphocyte cell count, and iii) lymphocyte cell depletion kinetics. A body-mapping tool permits recording the body locations for: a) physical dosimeters, b) erythema/wounds, and c)
radioactivity contamination. Five additional screens permit recording: 1) overview information, 2) patient‘s report, 3) exposure information, 4) radioisotope information, and 5) physician‘s notes. BAT also includes a summary report feature with the ability to output a report file compatible with the NATO Standardization Agreement (STANAG 2474 NBC/MED) entitled ―Determination and Recording of Ionising Radiation Exposure for Medical Purposes?. The application is extensively documented internally and can also serve as a training tool for use in a variety of radiological scenario exercises. BAT is distributed on-line upon review of a download request at website www.afrri.usuhs.mil. An alternative version of BAT (i.e., eBAT) distributed Medical Communications for Combat Casualty Care (MC4; website: www.mc4.army.mil/index.asp) with more secure data handling is also available.
4.3.2 First-responder Radiological Assessment Triage (FRAT)
The First-responders Radiological Assessment Triage (FRAT) is a software application being developed by the AFRRI to enable first responders to triage suspected radiation casualties based on the prodromal features listed in the Emergency Radiation Medicine Response?AFRRI Pocket Guide [AFRRI, 2008]. FRAT is designed for use on personal digital assistant (PDA) devices using the Palm operating system, and may eventually be available for other devices. FRAT was not designed to be a medical recording tool but rather a on the site analysis tool. With minimum text entry, FRAT provides screens to record and analyze: 1) signs and symptoms, 2) blood lymphocyte counts, and 3) dosimetry data. An expert panel provided input to permit weighing these multiple individual biodosimetric indices. The program will assess the multiparameter triage dose or the exposure without an assigned dose, or it will indicate there is no evidence of overexposure. Additional FRAT output features include triage dose-specific messages addressing 1) hematology guidance, 2) reliability and diagnostic information, 3) hospitalization estimations, and 4) mortality projections. The application can also serve as a training tool for use in a variety of radiological scenario exercises (Blakely et al., 2007).
4.3.3 Medical Decorporation (MEDECOR) Tool After a radiological dispersal device (RDD) event, it is possible for radionuclides to enter the human body through inhalation, ingestion, skin and wound absorption. From a health physics perspective, it is important to know the magnitude of the intake to perform dosimetric assessments. From a medical perspective, removal of radionuclides leading to dose aversion is of high importance. The efficacy of medical decorporation strategies is extremely dependent upon the time of treatment delivery after intake. The ― golden hour?, or more realistically 3?4 hours, is imperative when attempting to increase removal of radionuclides from extracellular fluids prior to cellular incorporation. To assist medical first response personnel in making timely decisions regarding appropriate treatment delivery modes, it is desirable to have a software tool which compiles existing radionuclide decorporation therapy data and allows a user to perform simple diagnosis leading to potential appropriate decorporation treatment strategies. Sponsored by the Canadian Department of National Defence
(DND), the software application entitled MEDECOR (MEDical DECORporation) was developed. The MEDECOR tool was designed initially for use in either a PDA or laptop PC environment using HTML/Jscript to allow for ease of portability amongst different computing platforms. The target end users are trained EMS first responders, triage nurses and physicians. The software falls under the categories of both medical triage and medical treatment. The availability is through the Department of National Defence (CANADA).
4.3.4 Internal Dosimetry
The Integrated Modules for Bioassay Analysis (IMBA, 2005) is a software application that allows a user to perform detailed internal dosimetry assessments, both dose calculations and bioassay. The bioassay module allows a user to define an intake regime (inhalation, ingestion, injection, wound, or vapour), timeframe (acute
or chronic) and radionuclide/intake (activity), and is capable of generating bioassay data (for example,
excretion in urine, feces, etc) and effective dose. An alternate calculation mode allows the user to specify
bioassay data (as may be available forensically after an RDD event) and estimate intake (hence dose). IMBA
is a simple to use GUI oriented application, although departure from default model parameters requires the
user to have expertise with internal dosimetry. IMBA Professional Plus is available at
http://www.imbaprofessional.com.
4.4 Summary
The potential impact of a radiological or nuclear (RN) event can place on government services, first responders and hospitals can be overwhelming. It is therefore of vital importance to ensure rapid response at all levels. The advent of small and powerful computing platforms in the last decade have made it possible to take rich health physics and medical data that only existed in reach-back facilities and bring it forward to the incident responders. Furthermore, reach-back centers and client-server software tools are making it easier to access even more advance software tools and databases when more detailed analysis and expertise is needed for major planning or response activities. The list of software programs presented here is not exhaustive, but does provide a representation of the depth and breadth of tools available.
5.0 DEVELOP INJURY ASSESSMENT AND BIOLOGICAL DOSIMETRY GUIDES
AND DOCUMENTS [GBR, USA]
There was interest among the working group members to assist in the revision and development of documents for Injury Assessment and for Biological Dosimetry. Members of the working group specifically contributed to the revision of AMedPC-6, AMedPC-8, and most recently in the development of the World Health Organization‘s new BiodoseNet program. Through networking, this small working group of experts has
established a continuum of reach-back advisory capabilities that have been called into action to support
various national and international NATO activities.
6.0 DEVELOP A PROPOSAL FOR ADVANCED DEMONSTRATION
TECHNOLOGY BY THE FINAL RTG-099 MEETING [CAN, CZE, DEU, FRA, GBR, POL, SWE, USA]
In 2007, NATO funded a new project entitled ―Development and Field Testing of a System for ―Alternate
Retrospective Radiation Dosimetry Methods to Aid in Response to an R/N Terrorist Incident?. The overall objective of the project is to develop and field-trial a system with associated protocols to provide standardized rapid dose assessments for mass-casualty management employing both biodosimetry and luminescence dosimetry (OSL) methods. This three-year (2008?2010) project addresses the final task of the working group that was aimed at developing a proposal for Advanced Demonstration Technology.
7.0 CONCLUSIONS
Radiation Injury Assessment and Biodosimetry Subgroup #2 was successful in identifying a Program of Work and outlining specific tasks to be addressed during the course of three years. All these tasks were successfully
addressed by the Working Group and presented in this summary report. Accomplishments of this project
included:
1. A prototype tool for selection, advancement, implementation and validation of complementing biodosi-metry assays;
2. A report on validated information for lead candidate multi-parameter biodosimetry tools;
3. A review of mi l i tary relevant software applications designed to provide radiation injury assessment,
biological dosimetry information and guidance to the first-responders and first-receivers;
4. Assistance in the development of injury assessment and biological dosimetry guides and documents;
5. A proposal for the development of Advanced Demonstration Technology to be validated through field
trials;
6. Identification of new gaps and directions for future activities to enhance NATO capabilities in managing Radiological/Nuclear casualties.
This NATO Human Factors and Medicine Working Group offered an opportunity for international partner-ships and for leveraging of national activities to contribute and enhance developments at an international
level. It was this working group‘s opinion that the final outcomes had a significant impact on both national and international development of capabilities and that these efforts need to be continued and supported in the future.
ACKNOWLEDGEMENT
The views expressed here are those of the authors; no endorsement by the U.S. Department of Defense has been given or inferred. AFRRI supported this research under work unit RAB4AL. The authors wish to thank
the AFRRI staff members William E. Dickerson, and John R. Mercier for their contributions.

REFERENCES


10. 2012年10月03日 01:40:02 : jDO82o1xf2
やはり、物理的に成型する時間は取れないので、
抽出データのみ貼り付けます。

11. 2012年10月03日 01:42:38 : jDO82o1xf2
Chapter 7
Early Biodosimetry Response:
Recommendations for Mass-Casualty Radiation Accidents and Terrorism*

W.F. Blakely
Armed Forces Radiobiology Research Institute
Uniformed Services University
8901 Wisconsin Avenue, Bethesda, MD, 20889-5603


The accepted generic multiparameter and early-response approach includes measuring radioactivity and monitoring the exposed individual; observing and recording prodromal signs/symptoms and erythema;
obtaining complete blood counts with white-blood-cell differential; sampling blood for the chromosome-aberration cytogenetic bioassay using the “gold standard” dicentric assay (translocation assay for long times
after exposure) for dose assessment; bioassay sampling, if appropriate, to determine radioactive contamination;
and using other available dosimetry approaches. In the event of a radiological mass-casualty incident, local,
national and international resources need to be integrated to provide suitable dose assessment and continuing clinical triage and diagnoses. This capability should be broadly based and include i) training and equipping local responders with tools and knowledge to provide early radiological triage, ii) establishing radiological teams capable to rapidly deploy and provide specialized dose assessment capabilities (i.e., radiation screening and radiobioassay sampling, hematology, etc.), and iii) access to reach-back expert reference laboratories (i.e., cytogenetic biodosimetry-, radiation bioassay-, electron paramagnetic resonance-based dose assessment). This multifaceted capability needs to be integrated into a biodosimetry “concept of operations” for use in a mass-casualty radiological emergency. On-going research efforts to identify and validate candidate screening and triage assays should ultimately contribute towards approved, regulated biodosimetry devices or diagnostic tests integrated into local, national, and international radioprotection programs.
1.0 INTRODUCTION
The effective medical management of a suspected acute-radiation overexposure incident necessitates recording dynamic medical data, measuring appropriate radiation bioassays, and estimating dose from dosimeters and radioactivity assessments in order to provide diagnostic information to the treating physician and a dose assessment for personnel radiation protection records. The accepted generic multiparameter and early-response approach includes measuring radioactivity and monitoring the exposed individual; observing and recording prodromal signs/symptoms and erythema; obtaining complete blood counts with white blood
cell differential; sampling blood for the chromosome-aberration cytogenetic bioassay using the "gold

*
Manuscript adapted from prior publication, Blakely WF (2008) Early Biodosimetry Response: Recommendations for Mass
Casualty Radiation Accidents and Terrorism. Refresher course for the 12th
International Congress of the International Radiation
Protection Association, October 19?24, 2008, Buenos Aires, Argentina, accessible at web site:
http://www.irpal2.org.ar/PDF/RC/_12_fullpaper.pdf
standard" dicentric assay for dose assessment; bioassay sampling, if appropriate, to determine radioactive contamination; and using other available dosimetry approaches (e.g., dose assessment by measurement of free radicals in solid matrix materials using electron paramagnetic resonance, EPR) [1?2]. The practice of radiation medicine dictates the establishment of response capability for rapid medical diagnosis and management of individuals overexposed. For example, many nations have established reference expert cytogenetic biodosimetry laboratories.
There are hundreds of instances in which one or more persons were accidently overexposed to ionizing radiation [3]. A subset of these incidents involves mass-casualty scenarios [2, 4]. A radiological or nuclear attack is also a possibility [5]. Because of recent terrorist activities and intelligence information, there is strong sentiment that it
is not a question of if, but when, a radiological or nuclear terrorist attack will occur [6].
The clinical medical decision needs associated with potential mass-casualty events prompted Lloyd and colleagues to advocate the diagnostic role of cytogenetics in early triage of radiation casualties [7]. Reference expert cytogenetic laboratories have recently established regional (e.g., reference cytogenetic laboratories among the nations of the United Kingdom, Germany, and France) and national [8?10] networks to enhance
their capabilities. In cases of urgent need for assessment in radiological exposures, individual nations often rely on international cooperation facilitated by United Nation (UN) agencies (i.e., World Health Organization
or WHO, International Atomic Energy Agency or IAEA). In the event of a radiological mass-casualty incident, current national and international resources need to be enhanced to provide suitable dose assessment and medical triage and diagnoses.
A coordinated approach involving preplanning, stockpiling of reagents and equipment, establishment and exercise of specialized response teams, and a consensus ―concept of operations? for biodosimetry applications in a mass-casualty radiological incident is required. Proper equipment for identifying radiation and radioactive contamination needs to be available to trained first responders. Specialist in radiological protection must also be available to provide expert advice and assistance to implement critical operational biodosimetry functions [11]. Preplanning and stockpiling of suitable reagents and equipment are essential [12]. Consensus concept-of-operations for biodosimetry application during a mass-casualty radiological emergency, tailored for specific radiological scenarios (i.e., radiation dispersal device, radiation emitting device, and improvised nuclear device), are also needed. The United States Office of Science and Technology Policy and the Homeland Security Council established an interagency working group that prioritized research areas for radiological nuclear threat countermeasures including efforts to automate biodosimetric assays and develop
biomarkers for biodosimetry [13]. These biodosimetric research efforts are focused to identify, optimize, and validate novel biodosimetric assays to support triage, clinical, and definitive radiation dose and injury.
2.0 BIODOSIMETRY PREPLANNING
2.1 Radiation exposure assessment methods
Table I illustrates a list of radiation exposure assessment methods applicable for early-phase acute radiation
based on international consensus of experts [2]. Protocols for use of these established and provisional methods
were also described in the Appendix section of this report. Table I also shows several features associated with
these assessment methods for considering their use for early triage screening, applicability for scoring acute
radiation syndrome (ARS) severity, and criteria for their use to prioritize suspected exposed individuals dose
assessment by cytogenetic biodosimetry. Depending on the radiation scenario and available resources,
appropriate radiation assessment methods should be implemented in a mass-casualty radiological terrorism or
radiation accident incident.
Table I: Acute-phase patient assessment methods*
* The table was modified from a version reported by Alexander and colleagues [2]. Note that the personal and area

monitoring methods are listed in alphabetical
order and, therefore, their location in the table does not infer priority or preference.
# Radiobioassay detection limits and costs are based on
137
Cs isotope and 1 min gamma-ray spectrometry analysis with high priority count (costs 3-times
routine) with no automatic sample changers used. Detection limits for cytogenetic analysis are presented in acute photon

equivalent dose in units of Gy.

Assessment Method
Parameters for considering assessment method
for use in early ( 25 min ? ?
Personal monitoring (indirect, invasive)
Detection
limit
Estimate cost per
sample, US Dollars#

blood chemistry (i.e., amylase activity) 1 d 50 pCi/swab $70 ?
stool sample > 1 d 5 pCi/g $80 ?
urine sample (spot; 24-hr) 1 d 30 pCi/vial $90 ?
cytogenetics (i.e., 20?50 metaphase triage;
1000 metaphase analysis)
> 3 days
1 Gy;
0.2 Gy
Unknown; $500?3,000 ?
Area monitoring
dosimetry results (e.g. TLDs, aerial measurements)
combined with personal location information
Unknown ? 3?7
2.2 Radiation/radiological response teams and networks
Specialist in radiation protection supporting early-response to radiation emergencies are typically organized
into teams with discrete functions as illustrated in Table II. For example, Remick and colleagues [14]
described U.S. national resources of response teams for radiological incidents. In certain nations,
components of these radiological resources are organized into teams that address assessment and medical
response for nuclear, biological, and chemical threats [15].

Table II: Selected List of Radiological Response Teams
Initial Assessment Nuclear, Chemical, and Biological
Radiation Source Search Medical Recording and Registry
Radiation Survey and Bioassay Sampling Haematology and Cytogenetic Biodosimetry Sampling

Specialized radiation teams are accessible through United Nation agencies. In 2000 IAEA established the
―Response Assistance Network? or RANET [16], previously called Emergency Response Network
(ERNET), of teams suitably qualified to respond rapidly and, in principal, on a regional basis, to nuclear or
radiological emergencies. RANET’s areas of assistance include: i) advisory, ii) assessment and evaluation,
iii) monitoring, and iv) recovery. WHO’s Radiation Emergency Medical Preparedness and Assistance
Network (REMPAN) [17] consists of biodosimetry laboratories with expertise in: cytogenetic, EPR,
bioassays, and molecular biology methodology. Recent efforts by WHO are focused to implement and
coordinate a global network of reference biodosimetry laboratories (Figure 1).


12. 2012年10月03日 01:46:27 : jDO82o1xf2
Figure 1: Illustration of networks of expert reference laboratories specializing in dose assessment by cytogenetic biodosimetry, electron paramagnetic resonance (EPR), molecular biodosimetry, and radioactivity measurements from biological samples or radiation bioassay
Koscheyev and colleagues [18] described that response teams responding to disasters can provide considerable benefits to both medical and psychological public-health problems. They also recommend use of a mobile diagnostic and a continuous operating pre-hospital triage system for rapid health screening of large populations after a large-scale disaster.
3.0 BIODOSIMETRY?CONCEPT OF OPERATIONS The primary purpose for early-response biodosimetry following suspected radiation overexposures is to rapidly provide first-responders and medical providers scientifically sound diagnostic radiation injury and dose assessment to support medical management treatment decisions. The measurement of clinical signs and symptoms associated with the severity of organ (i.e., hematological, gastrointestinal, neurovascular, and cutaneous) specific ARS, as developed and advocated by Prof. Fliedner (Ulm, Germany), is essential for triage of victims [19?20]. The risk of death from life-threatening radiation exposures is dependent on the level of medical care available (FIG. 2A). The U.S. Strategic National Stockpile (SNS) Radiation Working
Group [12] recommended a treatment approach using both the organ specific clinical signs and symptoms based on the Medical Treatment Protocols for Radiation Accident Victims (METREPOL) diagnostic system along with biological dosimetry (i.e., time to onset of nausea and vomiting, decline in absolute lymphocyte counts over several hours to days after exposure, and appearance of chromosome aberrations (i.e., dicentrics and rings). In the case of a mass casualty radiation emergency, this working group recommended cytokine,
antibiotic, and stem-cell transplant therapies, as illustrated in the dose windows shown in Figure 2B. The SNS Radiation Working Group also encouraged cytokine therapy to be initiated 24 h after radiation exposure, based on the preclinical studies by MacVittie and colleagues [21]. This will likely necessitate an initial reliance on diagnostic information based on early bioindicators of radiation dose, which will then be replaced by bioindicators of the severity of ARS response as the clinical case evolves.
The implementation of a multiparameter biodosimetry assessment approach is a significant confounder in a mass casualty radiological emergency. The U.S. Radiation Emergency Assistance Center/Training Site
(REAC/TS) has developed a ―Radiation Patient Treatment? treatment management approach that has incorporated early-response and multiple parameter biodosimetry when responding to a radiation incident with trauma or illness. The Armed Forces Radiobiology Research (AFRRI) has modified this approach, with permission from REAC/TS and incorporated it into AFRRI Pocket Guide ? Emergency Radiation Medicine Response (released July 2008); see website: www.afrri.usuhs.mil. Figure 3 illustrates the components of the REAC/TS and AFRRI treatment strategy along with the concept of operations for use of multiparameter biodosimetry A Joint Interagency Working Group (JIWG) under the auspices of the U.S. Department of Homeland Security Office of Research and Development conducted a technology assessment of emergency radiological dose assessment capabilities [22]. Gaps were identified to provide rapid radiation exposure triage. Current approaches and emerging technologies that offer potential to contribute in radiation injury and dose assessment response were identified. Research and development are needed to establish a diagnostic pyramid triage concept to facilitate a functional biodosimetry concept of operations in a mass-casualty radiation emergency (Figure 4). The initial screening radiation assay must be rapid (1 assay per minute or less), use a hand-held device, and ideally involve a self-use test. Secondary and tertiary radiation assay may require more expertise and take longer ( 2 Gy) and lasts only for a day or two. This information provides diagnostic information concerning partial- or whole-body exposures and can later help define the boundary of the radiation exposure area when skin graphs are necessary. The AFRRI Biodosimetry Worksheet (Appendix B) and BAT program
provide data templates for this purpose. The skin's response to radiation is biphasic, and this type of skin reaction is largely due to capillary dilation caused by the release of histamine-like substances. Erythema increases during the first week following exposure and then generally subsides during the second week. It may return 2?3 weeks after the initial insult and last up to 30 days, and additional changes, such as desquamation, bullae formation, or even skin sloughing may follow, all of which make even a crude estimation of radiation dose almost impossible.
4.2.3 Haematology
Haematological responses are an early response biomarker for radiation dose assessment and also contribute in the assessment of the severity of haematology ARS. Fliedner advocates the use of blood cell changes after whole-body radiation exposures are reliable bioindicators of injury and a critical aid to plan therapeutic treatments [31]. An approximate 50% decline occurs in peripheral blood lymphocyte counts over 12 hours that fall below normal vales (1.4 × 109/L) is indicative of a potential severe radiation overexposure [12].
Goans and colleagues introduced lymphocyte depletion kinetic models for dose estimates based on human radiation accident registry data for whole-body acute gamma exposures [28] and more recently for criticality accidents [29]. Immediately following exposure, a complete blood cell count (CBC) with white cell differential should be obtained and then taken three times a day for the next 2?3 days and twice a day for the following 3?6 days. The BAT program permits the recording of peripheral blood lymphocyte counts and then converts them into dose predictions using lymphocyte depletion kinetic models based on previous dose responses in radiation accidents [27, 28, 32]. Lymphocyte cell counts and lymphocyte depletion kinetics provide dose assessment predictions that fall in the equivalent photon dose range of 1?10 Gy; see Table III.

13. 2012年10月03日 01:48:41 : jDO82o1xf2
Table III: Biodosimetry Based on Acute Photon-Equivalent Exposures*

Dose
estimate
Time to Onset
of vomiting
Absolute lymphocyte count (×109/liter)
b (Day)
Lymphocyte
depletion
ratec

Relative increase
in serum amylase
activity at 1 d
compared with
normalsd


Number of dicentricse

Gy %a
Time (Hr) 0.5 1 2 4 6 8 Rate constant
Per 50
metaphases
Per 1000
metaphases
0 ? ? 2.45 2.45 2.45 2.45 2.45 2.45 ? 1 0.05?0.1 1?2
1 19 2.30 2.16 1.90 1.48 1.15 0.89 0.126 2 4 88
2 35 4.63 2.16 1.90 1.48 0.89 0.54 0.33 0.252 4 12 234
3 54 2.62 2.03 1.68 1.15 0.54 0.25 0.12 0.378 6 22 439
4 72 1.74 1.90 1.48 0.89 0.33 0.12 .044 0.504 10 35 703
5 86 1.27 1.79 1.31 0.69 0.20 0.06 .020 00.63 13 51 1034
6 94 0.99 1.68 1.15 0.54 0.12 0.03 .006 0.756 15
7 98 0.79 1.58 1.01 0.42 .072 .012 .002 0.881 16.5
8 99 0.66 1.48 0.89 0.33 .044 .006 < .001 1.01 17.5
9 100 0.56 1.39 0.79 0.25 .030 .003 < .001 1.13 18
10 100 0.48 1.31 0.70 0.20 .020 .001 < .001 1.26 18.5
* Table modified from version reported by Waselenko and colleagues [12]. Depicted above are the four most useful elements of biodosimetry. Dose range is based on acute photon-equivalent exposures. The first column indicates the percent of people who vomit, based on dose received and time to onset.
The middle left section depicts the time frame for development of lymphopenia. Two or more determinations of blood lymphocyte counts are made to predict a rate constant which is used to estimate exposure dose. The middle right section shows the relative increase in serum amylase activity in humans 1 day after radiation exposure. The final column represents the current ―gold standard? which requires several days before results are known. CSF
therapy should be initiated when onset of vomiting, lymphocyte depletion kinetics, and/or serum amylase suggests an exposure dose for which treatment is recommended. Therapy may be discontinued if results from chromosome dicentrics analysis indicate lower estimate of whole-body dose.
a.
Cumulative percentage of victims with vomiting.
b.
Normal range: 1.4?3.5 × 109
/L. Numbers in bold fall within this range.
c.
The lymphocyte depletion rate is based on the model Lt = 2.45 × 109/L × e-k(D)t where Lt equals the lymphocyte count (×109/L), 2.45 × 109
/L equals a constant representing the consensus mean lymphocyte count in the general population, k equals the lymphocyte depletion

rate constant for a specific acute photon dose, and t equals the time after exposure (days).
d.
Relative increases in serum amylase activity compared with normal [42].
e.
Number of dicentric chromosomes in human peripheral blood lymphocytes.


14. 2012年10月03日 01:53:09 : jDO82o1xf2
4.2.4 Blood chemistry assay

Blood biochemical markers of radiation exposure have also been advocated for use in early triage of radiation casualties [26, 33?35]. An increase in serum amylase activity (hyperamylasemia) from the irradiation of salivary tissue has been proposed as a biochemical measure of early radiation effect in a normal tissue [36?37]. Several studies have also advocated it use as a candidate biochemical dosimeter in man [26, 38?40]. A few hours after irradiation injury, cells in the salivary gland show acute inflammation and degenerative changes resulting in increases in serum amylase activity. Histochemical, isozyme analysis, and partial-body exposure studies confirm that the increase in serum amylase activity originated from the salivary glands.
Serum amylase activity increases occur early after head and neck irradiation of humans [41] and generally show peak values between 18?30 h after exposure, returning to normal levels within a few days [42].
Sigmoidal dose-dependent increases in the early (1 day) hyperamylasemia are supported by radio-iodine therapy [43?44], radiotherapy [38, 39, 45?46], and from limited data from three individuals exposed in a criticality accident [40]. Table III shows a dose response for relative increases in serum amylase activity 1 d after exposure. Significant inter-individual variations are reported in dose-response studies [37, 42, 45?46],
which represent a potential major confounder for use of serum amylase activity alone as a reliable biodosimeter. This inter-individual variation in biochemical response is not unexpected, since it is well known that the radiation level causing irreversible failure of the hematopoietic system varies among individuals and may reflect genetic and physiological differences and relative differences in the radiosensitivity of hematopoietic stem/progenitor cells [47] as well as radiation exposure parameters (i.e., partial-body exposures, shielding, dose-rate, etc.) [48].
4.2.5 Triage chromosome aberration cytogenetics
The quantification of the yield of chromosome aberrations (e.g., dicentric and rings) in lymphocyte metaphase spreads is one of the principal methods for estimating radiation dose to exposed individuals. The generation of
dose-response calibration curves is required for the proper evaluation of the lymphocyte-dicentric changes, which requires a high level of expertise and is typically performed in expert reference laboratories.
Dr. David C. Lloyd (National Radiological Protection Board, UK) suggested that cytogenetic triage using a lymphocyte metaphase-dicentric assay could be especially useful in providing evidence of non-uniform exposure and confirmation of individuals in a high-dose, exposed triage category [49]. In individuals with a high-dose estimate based on the mean number of dicentrics per cell, a significant fraction of metaphase spreads free of dicentrics suggests that surviving functional stem cells are present and that these individuals would be potential candidates for cytokine therapy versus a bone marrow transplant.
In managing radiation accidents, it is important to triage patients into broad, 1-Gy dose windows, especially when there are mass casualties and limited resources. Chromosome-aberration analysis using the conventional cytogenetic metaphase-spread dicentric bioassay is useful for the initial triage of mass casualties [4, 7, 12, 50?52]. In the triage mode only 40 to 50 metaphase spreads per subject are scored (fewer if the aberration yield is high) instead of the typical 500 to 1000 scored in a routine analysis. Table III demonstrates the expected triage (50 metaphases) and reference (1000 metaphases) yield of dicentrics for acute photon doses from 1 to 5 Gy.
After the initial results are communicated to the treating physician, additional scoring is advisable so that potential dose-assessment conflicts can be resolved, and assistance can be provided for physicians considering marrow stem cell transplants in high dose cases. Specialized cytogenetic biodosimetry laboratories need to develop and routinely practice emergency cytogenetic biodosimetry triage procedures.
4.2.6 Provisional and emerging triage, clinical, and definitive dose assessment methods
Several provisional and emerging approaches have been considered as methods to provide triage, clinical, and/or definitive dose assessment. For a review of these and other established dose assessment methods see reports by Alexander and colleagues [2] and Joint Interagency Working Group [22] and Table IV. Electron paramagnetic resonance (EPR)-based detection of free radicals is a well accepted and validated method for measurement of dose to dental enamel from biopsy teeth [2; 53] and has recently been extended to measure absorbed dose from teeth in vivo and nail clippings ex vivo [2]. Radiation causes injury to various tissues and organs resulting in time- and dose-dependent increases in blood. These proteins are bioindicators for radiation injury of relevant ARS organ systems (i.e., bone marrow, gastrointestinal system) as well as early bioindicators of absorbed dose. Gene array methods have been used to identify candidate radiation-response gene expression targets derived from blood lymphocytes and then measured by quantitative real-time reverse transcriptase polymerase chain reaction (QRT-PCR) methods. See Table IV below for a select listing and status of these provisional and emerging methods.

Table IV: Select list of provisional and emerging radiation injury and dose assessment methods
Method Status References
EPR
Teeth (in vivo)
EPR L-band is potentially able to measure doses as low as
2 to 3 Gy but needs additional development
2; 54?56
Nails (ex vivo) EPR X-band shows a lower limit of detection of 0.5?1 Gy 2; 57?59
Blood protein
immunoassay
C-reactive protein
Acute-phase reaction protein derived from liver and
demonstrated both as a biodosimeter and bioindicator of
hematology ARS
60?62
Flt-3 ligand Bioindicator of bone marrow injury 63?64
Citrulline Bioindicator of injury to small intestine epithelial tissue 65?67
γH2AX Protein associated with DNA double strand break repair 68
Multiple proteins
Candidate multiple protein biomarkers proposed for biodosimetry; multiple protein biomarkers demonstrated using multivariate discriminant or linear regression analyses methodology for radiation injury 69?70
Blood lymphocytes gene expression QRT-PCR assay of multiple targets
Multiple radiation responsive gene targets identified and used in the development of consensus dose-response calibration curves using an ex vivo blood radiation model system 71?75


5.0 RECOMMENDED BIODOSIMETRY ENHANCEMENTS FOR MASS-CASUALTIES RADIOLOGICAL INCIDENTS
In general first responders and medical care providers at hospitals have limited capabilities for assessing radiation injury, especially in the event of a radiological mass casualty incident. Further no single radiation bioassay at present is sufficient to provide robust dose-assessment capabilities for potential radiation exposure scenarios including mass casualties. A multiple parameter approach is necessary for triage, clinical, and definitive radiation biodosimetry [1]. A biological dosimetry approach for medically managing of a mass-casualty radiological emergency should include local, national, and international cooperation to: i) train and equip local responders with tools to provide early radiological triage capability, ii) establish deployable teams (equipped with hand-held and licensed devices to assess radiation exposure) and iii) access specialized reference laboratories (equipped with automation technologies to enhance their throughput).
Nations need access to expert radiobioassay, cytogenetic biodosimetry, molecular biodosimetry, and EPR dose assessment reach-back service laboratories. These capabilities provide nations with definitive dose assessment supporting radiation protection programs. Implementation of automation methods for these dose assessment methods have merit and should be linked with established service laboratories. Efforts to establish laboratory networks composed of national and international reference laboratories able to respond to a sudden surge of analysis requests from a mass-casualty incident should be encouraged and facilitated.
Radiological teams able to rapidly deploy to the emergency incident are essential to respond to mass casualty events and should follow guidelines recommended by IAEA’s RANET [16] and/or WHO’s REMPAN [17]
programs. These deployable teams capable of performing triage biodosimetry assays (blood cell counts, signs and symptoms assessments, and radioactivity biosampling) should also be exercised. The teams will need training [76] and be equipped with necessary supplies and equipment.
The development of biomarkers for biodosimetry has been identified as priority efforts to help the U.S.
prepare for the possibility of a terrorist attack using radiological or nuclear devices [12, 75]. Sustained research support is needed to identify, optimize, and validate novel hematological, cytological, and molecular radiation responsive biomarkers and biophysical dose assessment methods. The results from this effort should provide the basis for development of rapid and high-throughput applications leading towards licensed and effective hand-held and laboratory devices for assessing radiation exposure.
Finally, the countermeasures to enhance national and international medical response capabilities for radiological incidents, including mass casualty events, need to be integrated into the first-responder community ―all hazard? response concept in order to be effectively assimilated and sustained. Future hand-held and deployable laboratory devices used to measure radiation exposure based on a biological sample should have dual-use capabilities for assessing exposure to other threat agents (i.e., chemical and biological), which is consistent with an ―all hazard? approach. These research developments supporting biological dosimetry should be rapidly incorporated into local, national, and international ―radiological exercises? to enhance training for responders, and to increase knowledge of policy managers and the general public for the important role of biodosimetry in mass-casualty radiological emergencies.


15. 2012年10月03日 01:58:46 : jDO82o1xf2
ABSTRACT

This paper describes our efforts to automate blood-sample processing for cytogenetic preparations.
Cytogenetic analysis using short-term in vitro cultures from peripheral blood has a wide range of appli-cations including radiation biodosimetry, clinical pathology/molecular diagnostics, and genetic toxicology.
However preparation of cytogenetic specimens is elaborate and time consuming, and requires significant expertise and labour. To overcome these problems, a high-throughput robotic liquid-handler, along with automated cell harvester and spreader, were customized and integrated in an overall system. The system was capable of automated preparation of short-term in vitro blood cultures and metaphase spreads, with limited human interaction. To enhance quality control, instruments were interfaced with CytoTrack, a digital infor-mation management system for laboratory sample tracking and data management. We describe here the integration and customization of the equipment, and report a laboratory exercise leading to the preparation of about 100 metaphase spreads per day from whole-blood cultures. This exercise allowed us to estimate our capability to 500 metaphase spreads in a week for biodosimetry mass-casualty applications. Customization
and integration of laboratory automation equipment benefited cytogenetic analysis by increasing quality and throughput while decreasing work load.
1.0 INTRODUCTION
A very large number of assays with diagnostic, prognostic and discovery potential, including radiation bio-dosimetry, genetic toxicology and molecular diagnostics, relies heavily on cytogenetic analysis. Examples of such assays include the dicentric chromosome assay (DCA), the most recognized and internationally accepted ―gold standard? quantitative test for radiation-induced DNA damage and biodosimetry; fluorescent in situ hybridization (FISH), comparative genomic hybridization (CGH) and karyotyping, for the diagnosis and prognosis of genetic disorders and for cell-cycle related studies; micronucleus assay, part of a standard panel of tests for evaluation of genotoxicity and carcinogenicity of new drugs and chemicals prior to release for commercial use, and applicable to radiation biodosimetry [1?7]. For all these applications, sample processing is based upon short term cell culture in the presence of drugs affecting the cell cycle and upon preparation of spreads from swollen and fixed cells. Set up of blood cultures as well as preparation of high quality cytogenetic spreads, are time consuming, elaborate, and require highly trained and skilled personnel. Labour requirement for preparation and analysis of cytological specimens prohibits high-throughput production via manual means. Therefore, automation of sample processing and cytogenetic analysis is critical for large-scale applications. While automation for most cytogenetic applications has concentrated on data acquisition/
analysis and processing of tissue sections, we have devoted efforts to standardize a versatile, automated procedure for culture, fixation and analysis of metaphase spreads from HBPL (human blood peripheral lymphocytes). Our previous work concentrated on implementing a ―beta? version of sample-tracking system to eliminate data transcription error, increase efficiency and throughput, augment quality control and assurance, address data management and sample-tracking challenges likely to arise during sample processing and analysis after a radiation mass casualty [8]. Our recent work has focused on establishing a high-throughput cytogenetic protocol to process and analyze a large amount of samples for the dicentric assay. In this protocol, blood containing vacutainers are checked in a laboratory information management system (LIMS). A liquid-handling device is used to set up short term blood culture. Automated harvesters and spreaders are employed to produce high-quality metaphase spreads from cultured blood. Automated staining equipment is used for large-scale staining of slides, which are then analyzed with the help of a metaphase finder and multiple satellite chromosome-aberration analysis stations. This platform can be applied to (i)
generate a large number of high-quality chromosome metaphase spreads from human peripheral blood lymphocytes (HPBL), (ii) perform cytokinesis block for micronucleus assay, (iii) set up of standard as well as abbreviated blood cell culture techniques, such as interphase premature chromosome condensation (PCC) or rapid interphase chromosome analysis (RICA).
We describe here the challenges of integration and customization for high-throughput use and quality control of the equipment. Furthermore, we report a laboratory exercise for biodosimetry mass casualty applications leading to the preparation of about 100 metaphase spreads in one day, with an estimated production of 500 metaphase-spreads in a week from whole blood cultures.
2.0 METHODS
2.1 Blood collection and abridged cell culture procedure for the DCA
After obtaining informed consent, peripheral blood from healthy adult donors was collected by phlebotomy.
Two-hundred and fifty micro-liters of whole blood were added to 2.25 mL of MarrowMax culture bone marrow media (Gibco, USA) media premixed with 10 micro-liters/mL of PHA (Murex Diagnostics, UK).
Cells were stimulated to grow for 24 hours at 37°C. The cell cycle was arrested in first-division mitosis by the addition of colcemid followed by incubation for an additional 24 hours at 37°C. First-division metaphase spreads were harvested at 48 hours after culture initiation for chromosome aberration analysis [9].
2.2 Harvest/fixation/spreading/stain of metaphase spreads
An automated metaphase harvester (Hanabi PII, Chiba, Japan), was used to treat cells with hypotonic solution
(75 mM KCl) and fixative (ice-cold solution of 1:3 acetic acid:methanol) and to harvest metaphase spreads.
Following harvesting, 15-micro-liters of cell suspension (consisting of fixed and swollen cells) were dropped onto methanol cleaned, bar-coded glass slides using an automated spreader (HANABI P1 Metaphase
Spreader, Japan) and dried under controlled environmental conditions (37oC and 55% humidity). Two bar-coded slides were prepared from each sample (representing one patient) and Giemsa stained in a Thermo
Shandon Varistain Gemini autostainer (Thermo, US).
2.3 Process simulation and sample cross-contamination and sterility
Process simulation experiments were performed to optimize and test the Tecan robot’s functionalities using
PBS or FD&C Brilliant Blue 1 dye (McCormick & Company, MD). Undiluted FD&C Brilliant Blue 1 dye was aliquoted using the liquid handler. Tips were washed by aspirating and ejecting 30 mL of water, twice,
and then each tip was used to aliquot 100 micro-liters of PBS into a 96-well plate. The procedure was repeated 8 times. Sample cross-contamination was determined by spectroscopic reading at the max absorbance of the dye (630 nm).
2.4 Mycoplasma detection
The liquid handler was used to distribute fifteen 1-mL aliquots of sterile MarrowMax bone marrow medium in Falcon tubes. Aliquots were incubated at 37°C/5% CO2 for one week. Samples were tested for the presence of mycoplasma by PCR using the Maximbio Mycoplasma Detection PCR kit (cat no MP-70114), according to manufacturer recommendations. Samples were run on an agarose gel and stained with ethidium bromide.
2.5 Statistical analysis
Average and standard deviation were calculated in Excel. Percentage accuracy was expressed as percent error:
[average (observed)?average (expected)]/average (expected) × 100%. Measure of dispersion was calculated as coefficient of variation (CV) and was calculated by dividing the standard deviation by the mean value.
Criteria of acceptability were less than 5% percent error and less than 5% CV.
3.0 EQUIPMENT DESCRIPTION AND CUSTOMIZATION
Blood cultures were set up using the Tecan Freedom Evo 250 robotic liquid handler (Tecan, Switzerland), enclosed in a bio-safety level II hood (JT Baker Company, USA). Spreads were prepared from blood cultures via automated metaphase harvester and spreader (ADSTEC Corporation, Chiba, Japan). These instruments were interfaced with CytoTrack, a customized database used to overcome data management and sample tracking challenges. Several modifications were made to the liquid handling robot, harvester, and stainer to increase throughput and to provide additional quality control measures. Equipment description and custom-ization challenges are described below; the list of modifications made to the instruments is summarized in
Table I.
Table 1: Equipment customization and modifications for automating the preparation of metaphase spreads
Instrument Component Function Customization
Bio-safety enclosure
(BioProtect II, JT
Baker, USA)
Hood Sterility and worker safety Dimension to accommodate Tecan.
Tecan Freedom
Evo 250 (Tecan,
Switzerland)
Liquid Handler Arm Dispense liquid
Equipped with ceramic coated piercing tips
(Tecan, septa-piercing 5 ml tips, part no.
30025356).
Work deck
Worktable for labware
and reagents
Layout for whole-blood micro-culture.
Carrier for media
Hold Marrow-Max bone
marrow medium bottle
Custom made design and manufacture.

Racks for cell
culture tubes
Hold tubes
Equipped with adaptors (Tecan, cat no. 10619456) for tight fitting and with tape around the base.
Bar-code reader Positive chain-of-custody
Integrated with the Tube Position Validator
Service module and interfaced with Cytotrack.
Wash station
Wash tips between
aliquoting steps
Number and volume of washes
EVOware software Programming of Liquid Handler
Set up of an API interface with Cytotrack;
programming of custom made scripts; integration with the Tube Position Validator Service module.
Hanabi metaphase
harvester
Work deck
Loading and un-loading
of cell culture tubes
Equipped with temperature sensor probe.
Centrifuge Centrifugation Change rotor inserts.
Hanabi metaphase
spreader
Spreading surface
Spread metaphase
chromosomes under controlled
temperature and humidity
Equipped with temperature and humidity sensor probe.
3.1 Bio-safety level II enclosure A Class II, Type A2 bio-safety level II enclosure (BIOPROtect
R II, JT Baker Company, Maine, USA) was equipped with a HEPA-filtered air flow and used to host the robotic liquid handler. The height of the ceiling in the bio-safety hood was customized to accommodate the vertical movement of the liquid handler robot’s moving arm.
3.2 Bio-Tecan Freedom Evo 250 robotic liquid handler
The Tecan Freedom Evo 250 (Tecan, Switzerland), a robotic liquid handler run by the EVOWare software,
was equipped with a platform of approximate 200 cm in length, a robotic arm for liquid-handling (LiHA), a large customized work deck, a module for barcode identification of samples and labware, racks and carriers for reagents and vacutainers, and a wash station for pipette tips. To achieve high-throughput of sample processing, increase quality control and ensure safety for the operators, customizations were made to the liquid handling robotic arm (LiHA), and the work deck. New labware was custom made and EVOware programs were written to the set up and ensure quality control over whole-blood micro-cultures. The bar-code reader was integrated with a custom-made program and interfaced with a laboratory information management system, CytoTrack, to ensure maintenance of chain-of- custody.
3.2.1 Liquid Handler Arm (LiHA)
When manipulating blood, one of the main hazards is the potential contamination of the operator through aerosol or splashes. The LiHa was retrofitted with piercing tips, able to perforate the rubber caps on blood-containing vacutainers and to aspirate blood without the need to de-cap tubes, thus increasing throughput and safety. The rubber vacutainer caps presented significant resistance to piercing, causing the tips to bend and retract without perforating the cap. Several types of tips (both custom made and commercially available) had to be tested. Ultimately, a suitable off-the-shelf part was found.
3.2.2 Wash station
Between pipetting maneuvers, the LiHA washed the liquid handling tips in a designated wash station by aspirating/injecting water. In the automated cytogenetic system, two washes of 30 mL of liquid were per-formed for each tip during each cycle, to ensure lack of cross-contamination.
3.2.3 Bar-code reader and work deck A mobile barcode scanner, under the control of the EVOware software, was present on the work-deck and was set up to read barcodes on samples held in racks. For the automated cytogenetic protocol, the Tecan robot utilized the barcode scanner module along with the Tube Position Validator Service (TPVS), a custom-made program interfaced with EVOware that confirmed correspondence between vacutainer- and culture tube-barcodes prior to transferring of samples. If the correspondence between ―source? and ―destination? tubes was missing, the process was stopped and a warning message was displayed on the computer screen. A second program, TRIM (Tecan Robot Integration Module), parsed log file data from the Tecan and uploaded relevant data to the LIMS sample tracking database.
The work deck was set up to accommodate one carrier for cell culture media, and twelve sample-tubes racks,
each holding up to 16 tubes (Figure 1, Panel A). The cell culture media carrier (Figure 1, Panel B) was custom
made (Collins Welding, Inc) to hold three 200-mL bottles of media and three 20-mL bottles of PHA or colcemid. The carrier was 30-cm long (Y axis), 9.5-cm wide (X axis) and had a 3/8″ (9.5 mm) thick
aluminum base plate with cutout grooves conforming to Tecan work bench mounting clips. A 1″ (25.4-mm) thick black delrin body was placed on top of the plate.


16. 2014年5月29日 21:49:44 : Lo8eqPLQUA
De vives oppositions sont d窶冩res et dテゥjテ テ prテゥvoir entre les tenants d窶冰n porno テゥmancipateur de la femme, de sa sexualitテゥ, de l窶冩nanisme窶ヲ et ses pourfendeurs qui y voient bien plutテエt le symbole d窶冰ne <a href

  拍手はせず、拍手一覧を見る

この記事を読んだ人はこんな記事も読んでいます(表示まで20秒程度時間がかかります。)
★登録無しでコメント可能。今すぐ反映 通常 |動画・ツイッター等 |htmltag可(熟練者向)
タグCheck |タグに'だけを使っている場合のcheck |checkしない)(各説明

←ペンネーム新規登録ならチェック)
↓ペンネーム(2023/11/26から必須)

↓パスワード(ペンネームに必須)

(ペンネームとパスワードは初回使用で記録、次回以降にチェック。パスワードはメモすべし。)
↓画像認証
( 上画像文字を入力)
ルール確認&失敗対策
画像の URL (任意):
  削除対象コメントを見つけたら「管理人に報告する?」をクリックお願いします。24時間程度で確認し違反が確認できたものは全て削除します。 最新投稿・コメント全文リスト
フォローアップ:

このページに返信するときは、このボタンを押してください。投稿フォームが開きます。

 

 次へ  前へ

▲このページのTOPへ      ★阿修羅♪ > 原発・フッ素19掲示板

★阿修羅♪ http://www.asyura2.com/ since 1995
スパムメールの中から見つけ出すためにメールのタイトルには必ず「阿修羅さんへ」と記述してください。
すべてのページの引用、転載、リンクを許可します。確認メールは不要です。引用元リンクを表示してください。

     ▲このページのTOPへ      ★阿修羅♪ > 原発・フッ素19掲示板

 
▲上へ       
★阿修羅♪  
この板投稿一覧