22.12.2012 Views

Pan Arab Journal of Oncology - Arab Medical Association Against ...

Pan Arab Journal of Oncology - Arab Medical Association Against ...

Pan Arab Journal of Oncology - Arab Medical Association Against ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong><br />

ISSN: 2070-254X<br />

Official Publication <strong>of</strong> the <strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer | www.amaac.info | vol 3; issue 1 | March 10<br />

INITIATIVE TO IMPROVE CANCER CARE IN THE ARAB WORLD<br />

Proceedings <strong>of</strong> the Symposium<br />

March 23 - 25, 2010 | Riyadh, KSA


editorial board < contents <<br />

¼ Editor-in-Chief<br />

Marwan Ghosn, MD, MBA / MHM<br />

> mghosn.hdf@usj.edu.lb<br />

> marwan.ghosn@cmc.com.lb<br />

Lebanon<br />

¼ Deputy Editor<br />

Sami Khatib, MD<br />

> skhatib@khcbi.jo<br />

Jordan<br />

¼ Associate Editors<br />

Khaled Al-Saleh, MD<br />

> gffccku@yahoo.com<br />

Kuwait<br />

Jamal Khader, MD<br />

> jkhader@khcc.jo<br />

Jordan<br />

Hussein Khaled, MD<br />

> khaled@internetegypt.com<br />

Egypt<br />

Nazar Makki, MD<br />

> ntmakki@yahoo.com<br />

Iraq<br />

¼ Design & Layout<br />

ZŽ na Khairallah<br />

> zenakhairallah@gmail.com<br />

¼ PAJO Editorial Board<br />

> editorinchief.pajo@yahoo.com<br />

ISSN: 2070-254X<br />

<strong>Pan</strong> <strong>Arab</strong> Publishing Company<br />

P. O. Box: 2509<br />

Amman 11953 - Jordan<br />

Beer Al SabeÕ St<br />

Shocair <strong>Medical</strong> Complex<br />

2nd Fl, <strong>of</strong>fice No. 201<br />

Phone + 962 6 566 78 53<br />

Fax + 962 6 562 38 53<br />

www.e-pamj.com<br />

AMAAC Introduction > 2<br />

International Advisory Board > 3<br />

Editorial > 4<br />

Special Thanks > 5<br />

Cancer Care in the <strong>Arab</strong> World | March 23-25, 2010 | Riyadh, KSA > 6 - 93<br />

News from the <strong>Arab</strong> World > 94 - 100<br />

¼ The 5th SEMCO-ASCO Conference<br />

¼ First EURO-ARAB Congress<br />

¼ The 3rd Regional Congress <strong>of</strong> Cancer and Blood Disorders <strong>of</strong> Childhood<br />

¼ International Symposium on New Frontiers in Breast Cancer<br />

¼ Conference on Topics in Therapeutic and Diagnostic <strong>Medical</strong> Physics<br />

¼ The 10th <strong>Pan</strong> <strong>Arab</strong> Cancer Congress<br />

¼ The 7th International Jordan <strong>Oncology</strong> (JOS) Conference<br />

Cancer Awareness Calendar > 102<br />

Instructions for Authors > 103 - 106<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 1


amaac <<br />

AMAAC Introduction<br />

The <strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer (AMAAC) is a medical body that was established in 2001 as part <strong>of</strong> the <strong>Arab</strong> <strong>Medical</strong><br />

<strong>Association</strong> where its main <strong>of</strong>fice is located in Cairo - Egypt, and it is also a continuation <strong>of</strong> the <strong>Arab</strong> Council <strong>Against</strong> Cancer that<br />

was founded in 1995. The Executive Committee <strong>of</strong> (AMAAC) is represented by two members who are named <strong>of</strong>ficially by the<br />

<strong>Oncology</strong> Society <strong>of</strong> each <strong>Arab</strong> Country.<br />

The <strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer aims at strengthening relationships between members in different <strong>Arab</strong> Countries to<br />

raise the level <strong>of</strong> cooperation in the field <strong>of</strong> oncology on both scientific and practical aspects. Exchanging information and researches<br />

between members through Regional and <strong>Arab</strong> Conferences and Publications. Holding Public Awareness Campaigns in the field <strong>of</strong><br />

oncology that are organized by <strong>Arab</strong> Countries. Participating in scientific activities with International <strong>Oncology</strong> Societies. Finally,<br />

encouraging researchers and doctors to meet and exchange experiences together with finding training opportunities in the field <strong>of</strong><br />

oncology inside and outside the <strong>Arab</strong> World.<br />

> The Executive Board <strong>of</strong> AMAAC<br />

Sami Khatib, MD (Jordan) Secretary General<br />

Hussein Khaled, MD (Egypt) Associate Secretary General<br />

Maha Manachi, MD (Syria) Associate Secretary General<br />

Khaled Al-Saleh, MD (Kuwait) Associate Secretary General<br />

Brahim El Gueddary, MD (Morocco) Associate Secretary General<br />

Said Al-Natour, MD (Jordan) Associate Secretary General (for financial affairs)<br />

> The <strong>of</strong>ficially nominated members <strong>of</strong> AMAAC by the <strong>Oncology</strong> Societies <strong>of</strong> Each Country<br />

Algeria Adda Bounedjar, MD<br />

Kamel Bouzid, MD<br />

Bahrain Abdulla Ajami, MD<br />

Egypt Hussein Khaled, MD<br />

Sherif Omar, MD<br />

Iraq Abdul MonÕ em Ahmed, MD<br />

Nezar Taha Maki, MD<br />

Jordan Sami Khatib, MD<br />

Said Al-Natour, MD<br />

Kuwait Khaled Al Khalidi, MD<br />

Khaled Al Saleh, MD<br />

Lebanon Marwan Ghosn, MD<br />

Nagi El-Saghir, MD<br />

Libya Hussein A Hashemi, MD<br />

Rammah Rumaihi, MD<br />

Morocco Ashraki Abdel Kader, MD<br />

Brahim Khalil El Gueddari, MD<br />

Oman Bassim Bahrani, MD<br />

Palestine Fuad Sabatin, MD<br />

Abdel Razaq Salhab, MD<br />

Saudi <strong>Arab</strong>ia Om Al Kheir Abu Al Kheir, MD<br />

Shawki Bazarbashi, MD<br />

Sudan Hussein Mohammad Hamad, MD<br />

Kamal Eldein l Hamad, MD<br />

Syria Wassma Achawi, MD<br />

Maha Manachi, MD<br />

Tunisia Hamouda Boussen, MD<br />

Khalid Rahhal, MD<br />

Yemen Arwa Awn, MD<br />

Afif Nabhi, MD<br />

2 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


international advisory board <<br />

Matti AAPRO, MD<br />

Director, Multidisciplinary <strong>Oncology</strong> Institute, Genolier, Switzerland<br />

Consultant to the Scientific Director, European Institute <strong>of</strong> <strong>Oncology</strong>, Milano, Italy<br />

Consultant, Division <strong>of</strong> <strong>Oncology</strong>, Geneva University Hospital<br />

Geneva - Switzerland<br />

Hoda ANTON-CULVER, PhD<br />

Pr<strong>of</strong>essor & Chair<br />

Department <strong>of</strong> Epidemiology<br />

Pr<strong>of</strong>essor, Department <strong>of</strong> Microbiology and molecular Genetics,<br />

School <strong>of</strong> Medicine<br />

Director, Genetic Epidemiology Research Institute<br />

University <strong>of</strong> California<br />

Irvine – USA<br />

Jean-Pierre ARMAND, MD<br />

Pr<strong>of</strong>essor & General Director<br />

Centre de Lutte contre le Cancer<br />

Institut Claudius Regaud<br />

Toulouse – France<br />

Ahmad AWADA, MD<br />

Head <strong>of</strong> <strong>Medical</strong> <strong>Oncology</strong> Clinic<br />

Jules Bordet Cancer Institute<br />

Brussels - Belgium<br />

Patrice CARDE, MD<br />

Chairman Lymphoma Committee<br />

Gustave Roussy Institute<br />

Paris - France<br />

Franco CAVALLI, MD<br />

Pr<strong>of</strong>essor & President UICC<br />

Director<br />

<strong>Oncology</strong> Institute <strong>of</strong> Southern Switzerland<br />

Bellinzona - Switzerland<br />

Joe CHANG, MD<br />

Assistant Pr<strong>of</strong>essor <strong>of</strong> Radiation <strong>Oncology</strong><br />

Clinical Service Chief, Thoracic Radiation <strong>Oncology</strong><br />

MD Anderson Cancer Center<br />

Houston - USA<br />

William DALTON, MD<br />

President and Chief Executive Officer<br />

H.Lee M<strong>of</strong>fitt Cancer Center and Research Institute<br />

University <strong>of</strong> South Florida<br />

Florida - USA<br />

Jean-Pierre DROZ, MD<br />

Pr<strong>of</strong>essor & Former Head <strong>of</strong> <strong>Oncology</strong> Department<br />

Centre de Lutte contre le Cancer Leon Berard<br />

Lyon - France<br />

Alexander EGGERMONT, MD, PhD<br />

Pr<strong>of</strong>essor <strong>of</strong> Surgical <strong>Oncology</strong><br />

Head <strong>of</strong> Department <strong>of</strong> Surgical <strong>Oncology</strong><br />

Erasmus University <strong>Medical</strong> Center<br />

Daniel den Hoed Cancer Center<br />

Rotterdam - The Netherlands<br />

Jean-Pierre GERARD, MD<br />

Pr<strong>of</strong>essor <strong>of</strong> Radiation <strong>Oncology</strong><br />

General Director <strong>of</strong> Antoine-Lacassagne Cancer Center<br />

Lyon - France<br />

Joe HARFORD, MD<br />

Director <strong>of</strong> the Office <strong>of</strong> International Affairs<br />

National Institute <strong>of</strong> Health<br />

United States Department <strong>of</strong> Health and Human Services<br />

Bethesda - USA<br />

Alan HORWICH, MD<br />

Pr<strong>of</strong>essor <strong>of</strong> Radiotherapy<br />

Section <strong>of</strong> Academic Radiotherapy and<br />

Department <strong>of</strong> Radiotherapy<br />

The Institute <strong>of</strong> Cancer Research<br />

London – United Kingdom<br />

Fritz JANICKE, MD<br />

Director Clinic & Polyclinic <strong>of</strong> Gynecology<br />

University <strong>Medical</strong> Center Hamburg-Eppendorf<br />

Hamburg – Germany<br />

Sima JEHA, MD<br />

Director <strong>of</strong> the Leukemia / Lymphoma Developmental Therapeutics<br />

Saint-Jude Children’s Research Hospital<br />

Memphis - USA<br />

Hagop KANTARJIAN, MD<br />

Pr<strong>of</strong>essor <strong>of</strong> Medicine<br />

Chair <strong>of</strong> the Department <strong>of</strong> Leukemia<br />

The University <strong>of</strong> Texas - MD Anderson Cancer Center<br />

Houston - USA<br />

Fadlo R. Khuri, MD<br />

Pr<strong>of</strong>essor and Chair, Department <strong>of</strong> Hematology and <strong>Medical</strong> <strong>Oncology</strong><br />

Roberto C. Goizueta Distinguished Chair in Cancer Research<br />

Deputy Director, Clinical and Translational Research - Winship Cancer Institute<br />

Emory University School <strong>of</strong> Medicine<br />

Atlanta - USA<br />

Jean-Francois MORERE, MD<br />

Pr<strong>of</strong>essor at University Paris XIII<br />

Head <strong>of</strong> the Department <strong>of</strong> <strong>Oncology</strong><br />

Assistance Publique – Hôpitaux de Paris<br />

Paris - France<br />

Mack ROACH, MD<br />

Pr<strong>of</strong>essor & Chairman<br />

Radiation <strong>Oncology</strong> & Pr<strong>of</strong>essor <strong>of</strong> Urology<br />

University <strong>of</strong> California, Irvine<br />

California - USA<br />

Philippe ROUGIER, MD<br />

Pr<strong>of</strong>essor <strong>of</strong> <strong>Medical</strong> <strong>Oncology</strong><br />

Gastrointestinal Cancer<br />

Liver and <strong>Pan</strong>creas Tumors<br />

Ambroise-Pare Hospital<br />

Boulogne - France<br />

Youcef RUSTUM, PhD<br />

Chairman <strong>of</strong> the Department <strong>of</strong> Cancer Biology<br />

Roswell Park Cancer Institute<br />

Academic Research Pr<strong>of</strong>essor<br />

Associate Vice Provost<br />

University at Buffalo<br />

New York - USA<br />

Sandra M. SWAIN, MD<br />

<strong>Medical</strong> Director, Washington Cancer Institute<br />

Washington Hospital Center<br />

Washington – USA<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 3


editorial <<br />

Dear Colleagues,<br />

Despite considerable progress in its prevention and treatment, cancer remains a leading cause <strong>of</strong> morbidity and mortality in the <strong>Arab</strong><br />

World. Cancer Figures in our countries need action. Reasons are unknown. Costs <strong>of</strong> treatments are unaffordable. Healthcare system<br />

doesn’t <strong>of</strong>fer the optimal care. Patient and doctors are dissatisfied. The increase in medical tourism for a safer and less costly treatment<br />

is growing. Reports on the implementation <strong>of</strong> evidence-based medicine are few and even rare. The magnitude <strong>of</strong> the problem is not<br />

estimated, but we think that it is substantial. Challenges <strong>of</strong> the new environment are estimable.<br />

In its 1999 report Ensuring the Quality <strong>of</strong> Cancer Care, the Institute <strong>of</strong> Medicine's National Cancer Policy Board (NCPB) concluded,<br />

"Based on the best available evidence, some individuals with cancer do not receive care known to be effective for their condition." To<br />

address these concerns and the paucity <strong>of</strong> data on the quality <strong>of</strong> care for patients with cancer, the NCPB recommended establishing<br />

a quality monitoring system with capability <strong>of</strong> routine reporting <strong>of</strong> results.<br />

Motivated by the NCPB report, a number <strong>of</strong> initiatives were done to measure and improve the quality <strong>of</strong> care <strong>of</strong> patients with<br />

cancer. Examples include the joint effort <strong>of</strong> the National Cancer Institute and the National Quality Forum to identify and evaluate<br />

quality measures for cancer care; the American Society <strong>of</strong> Clinical <strong>Oncology</strong> Quality <strong>Oncology</strong> Practice Initiative; the <strong>Oncology</strong><br />

Demonstration Project sponsored by the Centers for Medicare and Medicaid Services; and the National Initiative for Cancer Care<br />

Quality (NICCQ), which is led by American Society <strong>of</strong> Clinical <strong>Oncology</strong> in collaboration with multiple oncology pr<strong>of</strong>essional<br />

societies, patient advocates, and researchers.<br />

We need to call for an urgent action to improve the Quality <strong>of</strong> care given to our patients. We need to optimize the treatment allocation,<br />

to ensure the access, the continuity <strong>of</strong> care and the integrity <strong>of</strong> the disease management with an acceptable cost effectiveness ratio.<br />

Changes <strong>of</strong> the healthcare system become a necessity and should face the challenges <strong>of</strong> the new environment. New solutions and<br />

strategies should be implemented.<br />

We are glad that the 1st issue <strong>of</strong> PAJO in 2010 is highlighting the initiative to improve Cancer Care in the <strong>Arab</strong> World. This meeting<br />

is hosted by National Guard Health Affairs jointly with <strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer in collaboration with national and<br />

international organizations and entities.<br />

We would like to thank all the contributors that worked hard to make this meeting happen.<br />

Marwan Ghosn, MD, MHHM<br />

4 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


special thanks <<br />

Thank you for all contributors, authors and reviewers <strong>of</strong> PAJO<br />

<strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> ournal <strong>of</strong> <strong>Oncology</strong><br />

<strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> ournal <strong>of</strong><br />

<strong>Oncology</strong> <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> ournal <strong>of</strong> <strong>Pan</strong> <strong>Oncology</strong> <strong>Arab</strong> <strong>Journal</strong> ournal <strong>of</strong><br />

<strong>Pan</strong> <strong>Oncology</strong> <strong>Arab</strong> <strong>Journal</strong> ournal <strong>of</strong><br />

<strong>Pan</strong> <strong>Oncology</strong> <strong>Arab</strong> <strong>Journal</strong> ournal <strong>of</strong><br />

<strong>Pan</strong> <strong>Oncology</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong><br />

Official Publication <strong>of</strong> the <strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer | www.amaac.info | vol 1; issue 2 | June 08<br />

ISSN: 2070-254X<br />

ISSN: 2070-254X<br />

ISSN: 2070-254X<br />

Official Publication <strong>of</strong> the <strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer | www.amaac.info<br />

| vol 2; issue 1 | January 09<br />

Official Publication <strong>of</strong> the <strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer | www.amaac.info<br />

Official Publication | vol <strong>of</strong> 2; issue the <strong>Arab</strong> 2 | <strong>Medical</strong> April 09 <strong>Association</strong> <strong>Against</strong> Cancer | www.amaac.info<br />

| vol 1; issue 3 | September 08<br />

Official Publication <strong>of</strong> the <strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer | www.amaac.info<br />

Official Publication | vol 2; issue <strong>of</strong> the 3 | <strong>Arab</strong> September <strong>Medical</strong> 09 <strong>Association</strong> <strong>Against</strong> Cancer | www.amaac.info<br />

Official Publication | vol 2; issue <strong>of</strong> the 3 | <strong>Arab</strong> December <strong>Medical</strong> 09<strong>Association</strong><br />

<strong>Against</strong> Cancer | www.amaac.info | vol 3; issue 1 | March 10<br />

Original Article<br />

Special Report<br />

Breast Cancer in Tunisia<br />

Highlights on the Speech and Language<br />

Special Issue Including the Proceedings <strong>of</strong> PACC 2009<br />

Pathologist’s role in Head and Health Neck Economics<br />

Cancer<br />

Review Articles<br />

Review<br />

A cost-minimization analysis Present & 9 Future <strong>of</strong> Radiation <strong>Oncology</strong><br />

S<strong>of</strong>t Tissue Sarcoma in Young Individuals MENA 2008<br />

<strong>of</strong> 1st line polyCT regimens in Review <strong>of</strong> the Current Management Management <strong>of</strong><br />

advanced NSCLC<br />

advanced advanced prostate cancer<br />

BLOM Beirut Marathon 08<br />

TH Special Issue Including the Proceedings <strong>of</strong> PACC<br />

Original Articles<br />

Review<br />

Meeting Highlights<br />

Treatment <strong>of</strong> Acute Lymphoblastic Leukemia ASCO 2008 Low dose Gemcitabine and Cisplatin<br />

PAN ARAB CANCER CONGRESS<br />

UICC 2008 in Advanced NSCLC<br />

Targeted Therapy Development<br />

PRAME and WT1 Genes expression<br />

7 - 9 May 2009 - Cairo, Egypt<br />

Angiogenesis review<br />

in CML Patients<br />

new publication<br />

Gerard Abadjian, MD<br />

Hamdi Abdel Azim, MD<br />

Wafaa Abdel-Hadi, MD<br />

A. Abdelkefi, MD<br />

Abdel Rahman M., MD<br />

Fatma Aboulkasem, MD<br />

Omalkhair Abulkhair, MD<br />

Mohsen Abdel Mohsen, MD<br />

<strong>Arab</strong>i Abdessamad, MD<br />

Noha Abdou, MD<br />

Miguel Aboud, MD<br />

Philippe Aftimos, MD<br />

Salim Adib, MD<br />

B. Allani, MD<br />

Bekadja Mohamed Amine, MD<br />

Elie Attieh, MD<br />

Fadwa Attiga, MD<br />

Ahmad Awada, MD<br />

Amal Baccar, MD<br />

Jean-Marc Bachaud, MD<br />

Thouraya Baroudi, MD<br />

Ali Bazerbachi, MD<br />

Amel Ben Ammar Elgaaied, MD<br />

Khaled Ben Rhomdhane, MD<br />

Alain Bernard, MD<br />

Ghislaine Bernard, MD<br />

Nizar Bitar, MD<br />

H. Boussen, MD<br />

Karim Chahed, MD<br />

Georges Chahine, MD<br />

Anouar Chaieb, MD<br />

Nicolas Chemali, MD<br />

Lotfi Cherni, MD<br />

Lotfi Chouchane, MD<br />

Elizabeth Cohen, MD<br />

Michel Daher, MD<br />

GŽ raldine Dalmasso, MD<br />

Kamal El-Dein Hamed Mohamed, MD<br />

new publication<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 5<br />

new publication<br />

new publication<br />

Dalia Darwish, MD<br />

Jean-Pierre Droz, MD<br />

Tayssir Eyada, MD<br />

Ahmad El-Ezzawy, MD<br />

Fadi Farhat, MD<br />

Nivine Gado, MD<br />

Marwan Ghosn, MD<br />

Heba Gouda, MD<br />

E. Gouider, MD<br />

Amin Haddad, MD<br />

Mohammad El-Hajj, MD<br />

Khaled Halahlah, MD<br />

Bechr Hamrita, MD<br />

Gregory Hangard, MD<br />

Colette Hanna, MD<br />

Mohamed A Hassan, MD<br />

Hassan A. Hatoum, MD<br />

Johan Hoebeke, MD<br />

Hesham El Hossieny, MD<br />

Ahmad Husari, MD<br />

Noha Ibrahim, MD<br />

Elias Jabbour, MD<br />

Sima Jeha, MD<br />

Maria Kabbage, MD<br />

Fadi El Karak, MD<br />

Joseph Kattan, MD<br />

M. Kefi, MD<br />

Jamal Khader, MD<br />

Hussein Khaled, MD<br />

Sami Khatib, MD<br />

Anne Laprie, MD<br />

Robert Launois, MD<br />

Katell Le Lay, MD<br />

Christelle Lemaitre-Guillier, MD<br />

Rami Mahfouz, MD<br />

Nazar Makki, MD<br />

Carole Massabeau, MD<br />

Andre Megarbane, MD<br />

While there’s<br />

there’s<br />

life,<br />

hope.<br />

(Cicero, 106 - 43 BC)<br />

Special Report: COMO 8 | Nov 2009 | Beirut, Lebanon<br />

Meeting Highlights<br />

InItIatIve to Improve CanCer Care In the arab World<br />

9th <strong>Pan</strong> <strong>Arab</strong> Original <strong>Oncology</strong> Articles Congress<br />

Effect <strong>of</strong> radiotherapy on malignant<br />

Best Best <strong>of</strong> ASCO Proteomic 2009 approach for the detection <strong>of</strong> pleural mesothelioma mesothelioma in in adjuvant,<br />

Proceedings <strong>of</strong> the Symposium<br />

breast cancer biomarkers.<br />

radical or palliative basis.<br />

March 23 - 25, 2010 | Riyadh, KSA<br />

new publication<br />

new publication<br />

Brahimi Mohamed, MD<br />

Mohsen Mokhtar, MD<br />

Walid Moukaddem, MD<br />

Jonathan Moyal, MD<br />

Elie Nasr, MD<br />

Fadi Nasr, MD<br />

Ghazi Nsouli, MD<br />

Ben Othman, MD<br />

Zaher Otrock, MD<br />

Martine Piccart, MD<br />

Shadi Qasem, MD<br />

Silvia Al Rabadi, MD<br />

Karim Rashid, MD<br />

Sami Remadi, MD<br />

Kamel Rouissi, MD<br />

Raya Saab, MD<br />

Ebtessam Saad El Deen, MD<br />

Laurence Ehret-Sabatier, MD<br />

Gamal Saied, MD<br />

Nagi El-Saghir, MD<br />

Ibrahim Saikali, MD<br />

Khaled El-Saleh, MD<br />

Ziad Salem, MD<br />

Lobna Sedky, MD<br />

Ali Shamseddine, MD<br />

Ahmad Shehadeh, MD<br />

Sana Al-Sukhun, MD<br />

Iyad Sultan, MD<br />

Ali Taher, MD<br />

Paul-Henri Torbey, MD<br />

Wafa Troudi, MD<br />

Virginie Vandenberghe, MD<br />

Alain Vergnenegre, MD<br />

Laure Vieillevigne, MD<br />

Besma Yacoubi-Loueslati, MD<br />

Mahmoud Yassein, MD<br />

Riad Younes, MD


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

Introduction<br />

This initiative aims at developing strategic recommendations to improve cancer<br />

care in the <strong>Arab</strong> countries.<br />

The inaugural meeting will be held on March 23-25, 2010 at the Four Seasons<br />

Hotel, Riyadh, KSA. This meeting is hosted by the National Guard Health Affairs<br />

jointly with <strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer in collaboration with national<br />

and international organizations and entities.<br />

In preparation to this meeting; working groups/ panels will be assembled to do<br />

the ground work and preparation for the launch <strong>of</strong> the initiative. The panels will<br />

have facilitators and members from various countries based on interest, expertise<br />

and nominations. While participants can hold an <strong>of</strong>ficial status in their countries,<br />

the participation in this event does not carry <strong>of</strong>ficial representation.<br />

The panels will work on performing situational analysis <strong>of</strong> a particular issue in<br />

the <strong>Arab</strong> World and suggest recommendations and specific action steps. These<br />

recommendations will be published and made available to all interested entities,<br />

organizations and individuals in the <strong>Arab</strong> Countries.<br />

Conference Objectives:<br />

1. To develop strategic recommendations to improve cancer care in the <strong>Arab</strong><br />

countries.<br />

2. To facilitate networking, experiences sharing, cooperation and collaborative<br />

projects across the <strong>Arab</strong> world.<br />

3. To recommend specific action steps pertinent to our countries in order to<br />

improve cancer care in the region.<br />

When: March 23 -25, 2010<br />

Where: Four Seasons Hotel<br />

Organizers:<br />

• National Guards Health Affairs, KSA<br />

• <strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer (AMAAC)<br />

Participating Organizations:<br />

• International Union <strong>Against</strong> Cancer (UICC)<br />

CANCER PREVENTION<br />

AND EARLY DETECTION<br />

• Saudi Cancer Society<br />

• Saudi Ministry <strong>of</strong> Health<br />

• <strong>Arab</strong>-European School <strong>of</strong> <strong>Oncology</strong><br />

• European Society <strong>of</strong> <strong>Medical</strong> <strong>Oncology</strong><br />

Sponsoring Organizations:<br />

• Roche<br />

• Novartis<br />

• Bayer<br />

• GE Health<br />

• Others<br />

Initiative to Improve Cancer Care in the <strong>Arab</strong> World<br />

The <strong>Oncology</strong> Department at National Guards Health Affairs, Riyadh, KSA and the<br />

<strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer (AMAAC) are launching the “Initiative<br />

to Improve Cancer Care in the <strong>Arab</strong> World (ICCAW)”.<br />

This initiative aims at developing strategic recommendations to improve cancer<br />

care in the <strong>Arab</strong> countries. The inaugural meeting will be held on March 23-25<br />

at the Four Seasons Hotel, Riyadh, KSA.<br />

For more information and registration in the meeting visit www.iccaw.com or<br />

email at info@iccaw.com.<br />

Abdul Rahman Jazieh, MD, MPH<br />

Chairman, Scientific Committee<br />

Initiative to Improve Cancer Care in the <strong>Arab</strong> World<br />

Dr. Sami Al Khatib<br />

Co-Chairperson, Scientific Committee<br />

Initiative to Improve Cancer Care in the <strong>Arab</strong> World<br />

Dr. Omalkhair Abulkhair<br />

Co-Chairperson, Scientific Committee<br />

Initiative to Improve Cancer Care in the <strong>Arab</strong> World<br />

6 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


MANUAL OF CANCER PREVENTION AND EARLY DETECTION<br />

GUIDELINES IN THE ARAB COUNTRIES<br />

Editors<br />

Abdul Rahman Jazieh, MD, MPH<br />

Chairman, Department <strong>of</strong> <strong>Oncology</strong><br />

King Abdulaziz <strong>Medical</strong> City<br />

Dr. Omalkhair Abulkhair<br />

Section Head, Division <strong>of</strong> Adult <strong>Medical</strong> <strong>Oncology</strong><br />

Department <strong>of</strong> <strong>Oncology</strong><br />

King Abdulaziz <strong>Medical</strong> City<br />

Introduction<br />

The Manual <strong>of</strong> “Cancer Prevention and Early Detection Guidelines in the <strong>Arab</strong><br />

Countries” is a compilation <strong>of</strong> manuscripts written by experts and physicians<br />

who participated in the Second Cancer Prevention and Early Detection Day held<br />

by National Guards Health Affairs in Riyadh, KSA on October 22 -23, 2008.<br />

The manuscripts review the epidemiological data and risk factors for common<br />

cancer and discuss risk factors, prevention and early detection. The authors<br />

presented the best guidelines and recommendations available for our <strong>Arab</strong><br />

Countries. It is known that there is a knowledge gap in this matter in term <strong>of</strong><br />

having original, accurate and adequate information from our region. Nevertheless,<br />

initial steps to combat cancer should be taken while more specific data relevant<br />

to our region made available.<br />

We thank all the authors for their efforts and contribution and we encourage all<br />

physicians to take part in the fight against cancer which will be the epidemic <strong>of</strong><br />

the 21st century.<br />

Abdul Rahman Jazieh, MD, MPH<br />

Chairman, Department <strong>of</strong> <strong>Oncology</strong>, King Abdulaziz <strong>Medical</strong> City<br />

Omalkhair Abulkhair, MD<br />

Section Head, Division <strong>of</strong> Adult <strong>Medical</strong> <strong>Oncology</strong><br />

Department <strong>of</strong> <strong>Oncology</strong>, King Abdulaziz <strong>Medical</strong> City<br />

Topics Index<br />

I. Cancer Epidemiology<br />

1. Epidemiology <strong>of</strong> Cancer in the Gulf Region<br />

2. Cancer Magnitude in Sham Countries<br />

II. Cancer Risk Factors<br />

1. Role <strong>of</strong> Diet in Cancer<br />

2. Oncologist Perspective on Tobacco Control: Historical View and Practical<br />

Guidelines<br />

3. Environmental Carcinogens and Pollution<br />

III. Breast Cancer<br />

1. Epidemiology, Prevention and Management Guidelines for Breast Cancer<br />

in <strong>Arab</strong> Countries<br />

2. The Role <strong>of</strong> MRI on Cancer Detection and Management: The Saudi <strong>Arab</strong>ian<br />

Experience<br />

3. Role <strong>of</strong> Imaging in Early Detection: Is Mammography Still the Standard<br />

<strong>of</strong> Care<br />

4. The Use <strong>of</strong> MRI for the Early Detection <strong>of</strong> Breast Cancer<br />

5. Diet, Physical Activity and Obesity in the Prevention and Recurrence <strong>of</strong><br />

Breast Cancer: Relevance to Saudi <strong>Arab</strong>ian Women<br />

6. Challenges <strong>of</strong> Cancer Screening Program<br />

IV. Gynecological <strong>Oncology</strong><br />

1. Endometrial Carcinoma<br />

V. GU <strong>Oncology</strong><br />

1. Urinary Bladder Cancer<br />

2. Testicular Cancer<br />

3. Prostate Cancer<br />

VI. Head and Neck Cancer<br />

1. Oral Malignancies: Role <strong>of</strong> Prevention and Early Detection<br />

2. Thyroid Cancer Epidemiology and Prevention<br />

VII. Lung Cancer<br />

1. Lung Cancer Prevention and Early Detection<br />

VIII. GI Malignancies<br />

1. Esophageal Cancer<br />

2. Gastric Cancer<br />

3. Colorectal Cancer: Prevention and Early Detection<br />

4. Hepatocellular Carcinoma<br />

IX. Pediatric Malignancies<br />

1. Cancer Prevention and Early Detection in Pediatric Malignancies<br />

X. Role <strong>of</strong> Physicians in Cancer Prevention<br />

1. Role <strong>of</strong> Primary Care Physicians in the Fight <strong>Against</strong> Cancer<br />

2. Role <strong>of</strong> Primary Care Physicians in the Prevention and Early Detection <strong>of</strong><br />

Cancer: An Oncologist Perspective<br />

Program<br />

1. Plenary sessions 1-3: Will include presentations by world experts in the<br />

related topics. They will address the topic in global fashion and reflecting<br />

on its relevance to our region.<br />

2. Breakout sessions: Will include the panel members with the help <strong>of</strong> the<br />

international experts from in the particular topics. It will be a chance for<br />

personal interaction among the facilitators, experts and panel members. The<br />

breakout session activities will include:<br />

a. Agreement on the consensus recommendations.<br />

b. Planning the next action step to be achieved in the next 12 month.<br />

c. Review and update available resources list.<br />

A document should be generated from the meeting in uniformed publishable format.<br />

4. Plenary Session 4 - 5: The panels will present their recommendations and<br />

action steps to all attendees.<br />

5. Satellite Symposia: Will address specific clinical topics <strong>of</strong> interest to the<br />

practicing physicians in order to have valuable attractive educational events<br />

for attendees and will be organized by sponsors.<br />

6. Social activities: Opening Ceremony dinner will be held on March 23 night.<br />

Gala dinner will be held in March 24 night.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 7


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

EPIDEMIOLOGY OF CANCER IN THE GULF REGION<br />

Khoja, T. 1 , Zahrani A. 2<br />

(1) Council <strong>of</strong> Health Ministers for GCC State<br />

(2) Gulf Center for Gulf Registration<br />

Corresponding Author: Dr. Tawfik A. M. Khoja<br />

Family Physician Consultant, Director General, Executive Board, Council <strong>of</strong><br />

Health Ministers for GCC State, P.O. Box 54647 Riyadh 11524, Kingdom <strong>of</strong><br />

Saudi <strong>Arab</strong>ia<br />

E-mail: sgh@sgh.org.sa<br />

Introduction<br />

Cancer is the second most frequent cause <strong>of</strong> death in majority <strong>of</strong> the developed<br />

countries. It is emerging as a major public health problem in developing countries.<br />

The International Agency for Research on Cancer (IARC) estimates that, globally,<br />

nearly 11 million new cases <strong>of</strong> cancer and more than 6 million deaths from this<br />

disease occurred during 2002 with more than half <strong>of</strong> the cases arising in developing<br />

countries where resources for treatment and prevention are scarce. The rapid<br />

improvement in the field <strong>of</strong> health care together with the control <strong>of</strong> communicable<br />

diseases, increased life expectancy at birth, and with rapid socioeconomic changes<br />

resulting in modified lifestyles such as increased prevalence <strong>of</strong> tobacco use,<br />

decrease in physical activity and rapid uptake <strong>of</strong> unhealthy food habits has resulted<br />

in an increased incidence <strong>of</strong> cancer in the developing countries.<br />

Cancer registries are a unique source <strong>of</strong> information centre for any cancer control<br />

program. These data helps to allocate financial and manpower resources in cost-effective<br />

health care planning as well as in the design <strong>of</strong> early detection and prevention programs.<br />

The Gulf Center for Cancer Registration (GCCR) was established in 1997. The<br />

GCCR works under the jurisdiction <strong>of</strong> the Executive Office <strong>of</strong> the Health Ministers’<br />

Council for GCC States. The main <strong>of</strong>fice is located in the premises <strong>of</strong> the Research<br />

Center, King Faisal Specialist Hospital and Research Center. The GCCR database,<br />

population-based incidence data is the largest aggregations in the Asia. Compiling<br />

data from the six national cancer registries representing the six Gulf countries:<br />

United <strong>Arab</strong> Emirates (UAE), Kingdom <strong>of</strong> Bahrain, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

(KSA), Sultanate <strong>of</strong> Oman, State <strong>of</strong> Qatar, and State <strong>of</strong> Kuwait.<br />

Objectives<br />

The primary objective <strong>of</strong> this study is to classify information on all cancer cases in<br />

order to produce statistics on the occurrence <strong>of</strong> cancer among GCC States’ nationals.<br />

Materials and Methods<br />

Data which include patient’s identification, demographic information, site <strong>of</strong> cancer,<br />

histology, stage, behavior and extent <strong>of</strong> the disease, basis <strong>of</strong> diagnosis and treatment<br />

methods are collected from the patient’s medical records based on clinical and<br />

histological diagnosis at the National Cancer Registry (NCR) in each <strong>of</strong> the GCC<br />

States. Data entry in CanReg4 s<strong>of</strong>tware and data quality assurance are performed<br />

at each GCC State NCR before they sent to the GCCR main <strong>of</strong>fice for ensuring<br />

the accuracy <strong>of</strong> information reported and subsequently for annual data analysis.<br />

Crude Incidence Rates (CIR) and Age Standardized Rates (ASR) were calculated<br />

to compare cancer incidence between different countries. These rates are sensitive<br />

to changes in the number <strong>of</strong> reported cases due to underreporting and changes<br />

in the population structure. Therefore, there is a potential for some changes in<br />

cancer incidence rates in the GCC countries that did not have recent population<br />

census available for this report or those depend on fewer sources for cancer case<br />

identification.<br />

Overall cancer incidence in the GCC States<br />

From January 1998 to December 2004 there were 58,180 newly diagnosed cancer<br />

cases among the GCC States nationals reported from the six National Cancer<br />

Registries. 28,542 cases (49.06%) were males and 29,638 (50.94%) were females.<br />

Majority <strong>of</strong> cases (71.1%) were reported from KSA, followed by Oman (11.1%),<br />

Kuwait (6.8%), Bahrain (5.1%), UAE (3.8%), and Qatar (2.1%).<br />

Diagnosis <strong>of</strong> cancer based on histopathological confirmation in more than 95% <strong>of</strong><br />

cases ranging from 85.2% in Kuwait to 97.4% in KSA indicating high degree <strong>of</strong><br />

certainty <strong>of</strong> cancer diagnosis in trade<strong>of</strong>f completeness <strong>of</strong> cancer registration in the<br />

GCC States. Other sources such as clinical, surgical, and radiological reports were<br />

rarely used as independent sources for cancer registration. Death certificate was<br />

used as independent source for cancer registration by Kuwait (13.2%) and Bahrain<br />

(5.7%), whereas other countries showed no preference to this type <strong>of</strong> data collection.<br />

Almost half <strong>of</strong> the cancer cases had either regional or distant metastasis at the<br />

time <strong>of</strong> diagnosis. Only 22.6% <strong>of</strong> patients presented with localized tumors and<br />

less than 2% with in situ, indicating the necessity <strong>of</strong> launching early detection<br />

programs in the Gulf region. Unknown extent <strong>of</strong> cancers was reported in about<br />

28% <strong>of</strong> patients ranging from 11.1% in UAE to 73.8% in Bahrain.<br />

In males, cancer incidence was highest in Bahrain and Qatar nationals with ASR <strong>of</strong><br />

158.7 and 157 per 100,000 population for Bahraini and Qatari males, respectively.<br />

Kuwaiti males had an ASR <strong>of</strong> 132.6/100,000 population followed by Omani males<br />

with an ASR <strong>of</strong> 99.1/100,000 population, UAE males with ASR <strong>of</strong> 70.1/100,000<br />

population, and the lowest was among Saudi males (66.1/100,000 population).<br />

In females, cancer incidence was highest in Qatari women with ASR <strong>of</strong><br />

165.2/100,000 populations, followed by Bahraini women with ASR <strong>of</strong><br />

142.3/100,000 population, Kuwaiti women with ASR <strong>of</strong> 136.7/100,000 population,<br />

Omani women with an ASR <strong>of</strong> 85.4/100,000 population, UAE women with ASR<br />

<strong>of</strong> 81.9/100,000 population, and the lowest was among Saudi women (ASR<br />

62.9/100,000 population).<br />

Cancer is mainly the disease <strong>of</strong> old age. The mean age at diagnosis was 50.5 years<br />

(Standard Deviation ± 22.1). Total <strong>of</strong> 4029 cases (9.7%) were children below age<br />

<strong>of</strong> 15 years. Women develop cancer at an earlier age compared to men, the mean<br />

age for women was 47.7 years and 53.2 years for men. There is no significant<br />

difference in the age specific incidence rates in men and women <strong>of</strong> the GCC States.<br />

Most common cancers among nationals <strong>of</strong> the GCC States:<br />

Breast Cancer<br />

Breast cancer is the second most common cancer in the world and the most<br />

common cancer in women accounting to about 23% <strong>of</strong> all cancers. It is estimated<br />

that approximately one million cases <strong>of</strong> female breast cancer are diagnosed every<br />

year worldwide. Breast cancer is the most frequent cause <strong>of</strong> death among cancer<br />

deaths in women. An estimated 410,712 breast cancer deaths occurred in 2002.<br />

8 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


Breast cancer is the most common cancer in the GCC States. Between January<br />

1998 and December 2004, 6,882 breast cancer cases were reported from all GCC<br />

States accounted to 11.8% from all cancers and 22.7% from cancers among<br />

females. Bahrain reported the highest incidence <strong>of</strong> breast cancer. The ASR per<br />

100,000 women was 46.4 for Bahrain followed by Kuwait (44.3), Qatar (35.5),<br />

UAE (19.2), Oman (14.4), and KSA (12.9).<br />

Leukemia<br />

Leukemia is the 12th most common cancer in the world. The estimated number <strong>of</strong><br />

newly diagnosed cases <strong>of</strong> leukemia was 300,522 cases in 2002 accounting to about<br />

2.8% <strong>of</strong> all new cancer cases with male to female ratio <strong>of</strong> 1.32: 1. In 2002, there were<br />

222,506 reported deaths attributed to leukemia accounting to 3.3% <strong>of</strong> all cancer deaths.<br />

In the GCC States, leukemia is the second most common cancer. Between January<br />

1998 and December 2004 there were 4,890 cases <strong>of</strong> leukemia accounted to 8.4%<br />

from all cancers in the GCC States. Leukemia incidence appeared to be slightly<br />

higher in males than females. Lymphoid leukemia was more common in males<br />

while myeloid leukemia was more common in females.<br />

Non-Hodgkin’s Lymphoma (NHL)<br />

NHL is the 10th most common cancer in the world. There are 300,571 new cases <strong>of</strong><br />

NHL in 2002 accounting to 2.8% <strong>of</strong> all cancers. The male to female ratio is 1.39: 1. An<br />

estimated 171,820 deaths <strong>of</strong> NHL occurred in 2002 representing 2.6% <strong>of</strong> all cancer deaths.<br />

NHL is the third most common cancer in the GCC States. 4,830 NHL cases<br />

were reported from all GCC States accounted to 8.3% from all cancers between<br />

January 1998 and December 2004. NHL was the most common cancer in males<br />

and the third most common cancer in females (accounted to 10.4% and 6.3% for<br />

males and females respectively). Qatar reported the highest incidence <strong>of</strong> NHL<br />

among males and females (ASR was 11.8/100,000 for males and 8.0/100,000 for<br />

females), followed by Kuwait (ASR was 11.0/100,000 for males and 6.8/100,000<br />

for females). Oman and Bahrain males ranked third with ASR <strong>of</strong> 7.8/100,000,<br />

whereas Bahraini women ranked third and Kuwaiti women ranked fourth with<br />

ASR 5.4/100,000 and 4.4/100,000 respectively. KSA and UAE women ranked<br />

fifth with ASR <strong>of</strong> 4.1/100,000 each. KSA men ranked fifth and UAE men ranked<br />

sixth with ASR <strong>of</strong> 5.6 and 5.2 per 100,000 populations respectively.<br />

Colorectal Cancer<br />

Colorectal cancer is the third most common cancer in the world. In 2002, there<br />

were approximately one million new cases <strong>of</strong> colon and rectum cancer, 550,465<br />

males and 472,687 females worldwide. Colorectal cancer incidence ranked fourth<br />

in men and third in women. Approximately there were 529,000 deaths attributed<br />

to colon cancer in 2002.<br />

Colorectal cancer is the fourth most common cancer in the GCC States. 4,213<br />

colorectal cancer cases (7.2% from all cancers) were reported from all GCC<br />

States between January 1998 and December 2004. Kuwait reported the highest<br />

incidence <strong>of</strong> colorectal cancer (ASR was 15.1/100,000 population) among males<br />

followed by Bahrain (12.6/100,000), Qatar (11.6/100,000), UAE (6.6/100,000),<br />

KSA (5.0/100,000), and Oman (4.5/100,000), while Qatar reported the highest<br />

incidence <strong>of</strong> colorectal cancer (14.1/100,000) among females followed by Kuwait<br />

(12.7/100,000), Bahrain (7.3/100,000), UAE (5.3/100,000), KSA (4.7/100,000)<br />

and Oman (3.8/100,000).<br />

Thyroid Cancer<br />

Thyroid cancer is the 21st most common cancer in the world. The estimated total<br />

number <strong>of</strong> new thyroid cancer cases was 141,013 with an estimated 35,375 deaths<br />

from thyroid cancer representing 0.53% <strong>of</strong> all cancer deaths in 2002. Thyroid<br />

cancer is almost 3 times higher in females compared to males.<br />

Thyroid cancer is the fifth most common cancer in the GCC States. 3,458 thyroid<br />

cancer cases (5.9% from all cancers) were reported from all GCC States between<br />

January 1998 and December 2004. Thyroid cancer incidence was significantly<br />

higher among women compared to men in all <strong>of</strong> the GCC States. It ranked second<br />

most common cancer in women next to breast cancer. Qatar reported the highest<br />

incidence with ASR <strong>of</strong> 13.5/100,000 followed by Kuwait (7.7/100,000), Bahrain<br />

(7.6/100,000), UAE (6.0/100,000), Oman (5.9/100,000), and KSA (5.0/100,000).<br />

Liver Cancer<br />

Liver cancer is the sixth most common cancer in the world, with an estimated<br />

number <strong>of</strong> 626,162 (5.8% <strong>of</strong> new cancer cases) in 2002. Males are affected more<br />

than females with male to female ratio <strong>of</strong> 2.40: 1. Because <strong>of</strong> the very poor<br />

prognosis, number <strong>of</strong> deaths in 2002 (598,321) was not far short <strong>of</strong> the number <strong>of</strong><br />

new cases, and it represents the third most common cause <strong>of</strong> death among cancer<br />

deaths accounting to 8.9% <strong>of</strong> cancer deaths.<br />

Liver cancer is the sixth most common cancer in the GCC States. 2,987 liver cancer<br />

cases (5.1% from all cancers) were reported from all GCC States between January<br />

1998 and December 2004. Liver cancer incidence was significantly higher among<br />

men compared to women in all <strong>of</strong> the GCC States. It ranked third most common<br />

cancer in men next to NHL and leukemia. Qatar had the highest incidence among<br />

men and women with ASR <strong>of</strong> 13.1/100,000 for males and 8.9/100,000 for females.<br />

Kuwaiti men ranked second (8.8/100,000), followed by KSA (6.9/100,000), Oman<br />

(6.4/100,000), Bahrain (5.2/100,000), and UAE (3.0/100,000).<br />

Lung Cancer<br />

Lung cancer is the commonest cancer in the world today accounting to 12.4% <strong>of</strong><br />

all new cases. There are estimated to be 1,352,132 new cases in 2002. The disease<br />

is more common in men with a male to female ratio <strong>of</strong> 2.49: 1. Lung cancer is the<br />

leading cause <strong>of</strong> cancer related death in men and second in women. An estimated<br />

1,178,918 deaths accounting to 17.5% <strong>of</strong> all cancer deaths are occurred in 2002.<br />

Lung cancer is the seventh most common cancer in the GCC States. 2,481 lung<br />

cancer cases (4.9% from all cancers) were reported from all GCC States between<br />

January 1998 and December 2004. Lung cancer incidence was significantly higher<br />

among men compared to women in all <strong>of</strong> the GCC States. It ranked fourth most<br />

common cancer in men next to NHL, leukemia, and liver cancer. Bahrain had the<br />

highest incidence among men and women with ASR <strong>of</strong> 34.2/100,000 for males<br />

and 12.0/100,000 for females. Qatar ranked second, followed by Kuwait, Oman,<br />

and UAE. KSA reported the lowest incidence among males and females (5.0 and<br />

1.3 per 100,000 for males and females respectively).<br />

Stomach Cancer<br />

Stomach cancer is the fourth most common cancer in the world with an estimated<br />

number <strong>of</strong> 933,937 cases in 2002 (8.6% <strong>of</strong> new cancer cases). The male to female<br />

ratio is 1.83: 1.<br />

An estimated 700,349 (446,052 males and 254,297 females) stomach cancer<br />

deaths occurred in 2002. It is the second most frequent cause <strong>of</strong> death among<br />

cancer deaths accounting to 10.4% <strong>of</strong> cancer deaths.<br />

Stomach cancer is the eighth most common cancer in the GCC States. 2,424<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 9


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

stomach cancer cases (4.1% from all cancers) were reported from all GCC States<br />

between January 1998 and December 2004. Stomach cancer incidence was more<br />

common in men compared to women in all <strong>of</strong> the GCC States. Oman had the<br />

highest incidence among men and women with ASR <strong>of</strong> 12.8/100,000 for males<br />

and 6.2/100,000 for females. Bahrain ranked second, followed by UAE, Qatar,<br />

and Kuwait. KSA reported the lowest incidence among males and females (2.8<br />

and 1.7 per 100,000 for males and females respectively).<br />

Prostate Cancer<br />

Prostate cancer is the fifth most common cancer in the world and the second<br />

importance in men. The estimated total number <strong>of</strong> new cases in 2002 is 679,023<br />

accounting to 11.7% <strong>of</strong> cancers in men. With an estimated 221,002 deaths prostate<br />

cancer is the sixth leading cause <strong>of</strong> cancer death in men. This represents 5.8% <strong>of</strong><br />

all cancer deaths in men.<br />

Prostate cancer is the 9th most common cancer in the GCC States. 1,721 prostate<br />

cancer cases were reported from all GCC States between January 1998 and<br />

December 2004 accounted to 3.0% from all cancers and 6.0% from cancers among<br />

males. Bahrain reported the highest incidence <strong>of</strong> prostate cancer. The ASR per<br />

100,000 men was 14.1 for Bahrain followed by Kuwait (12.3), Oman (9.6), Qatar<br />

(9.4), UAE (6.4), and KSA (4.0).<br />

Cervical Cancer<br />

Cancer <strong>of</strong> cervix is the second most common cancer in women in the world.<br />

Almost half a million newly diagnosed cases <strong>of</strong> cervical cancer are estimated<br />

every year accounting to about 10% <strong>of</strong> all cancers in women. Cervical cancer<br />

is the third most frequent cause <strong>of</strong> death among cancer deaths in women. An<br />

estimated 273,500 cervix cancer deaths occurred in 2002 accounting to about 9%<br />

<strong>of</strong> all cancer deaths in women.<br />

Cervical cancer is the 10th most common cancer in the GCC States. Between<br />

January 1998 and December 2004, there were 1,279 cervical cancer cases reported<br />

from all GCC States accounted to 2.2% from all cancers and 4.3% from cancers<br />

among females. Qatar reported the highest incidence <strong>of</strong> cervical cancer. The ASR<br />

per 100,000 women was 6.8 for Qatar followed by Bahrain (5.9), Oman (5.7),<br />

UAE (5.4), Kuwait (4.8), and KSA (2.2).<br />

Ovarian Cancer<br />

Ovarian cancer is the sixth most common cancer among women. In 2002, there<br />

were 204,499 newly diagnosed ovarian cancer cases accounting to 5.8% <strong>of</strong> all<br />

cancers among women. Ovarian cancer is also the seventh most common cause<br />

<strong>of</strong> death among cancer deaths in women. In 2002 there were 124,860 deaths<br />

attributed to ovarian cancer accounting to 4.3%.<br />

Ovarian cancer is the 11th most common cancer in the GCC States. Between<br />

January 1998 and December 2004, 1,180 ovarian cancer cases were reported<br />

from all GCC States accounted to 2.0% from all cancers and 4.0% from cancers<br />

among females. Bahrain reported the highest incidence <strong>of</strong> ovarian cancer. The<br />

ASR per 100,000 women was 7.4 for Bahrain followed by Qatar (6.7), Kuwait<br />

(5.9), Oman (5.5), UAE (4.4), and KSA (2.3).<br />

Conclusion<br />

The GCCR is a demonstration <strong>of</strong> the value <strong>of</strong> collaborative efforts among the<br />

region countries to establish epidemiological data about cancer that could be used<br />

in various ways in cancer control efforts.<br />

ROLE OF DIET IN CANCER<br />

Al-Sukhun, S. 1<br />

(1) University <strong>of</strong> Jordan, Amman<br />

Corresponding Author: Dr. Sana Al-Sukhun, MD, MSc<br />

Assistant Pr<strong>of</strong>essor <strong>of</strong> Medicine, Department <strong>of</strong> Hematology/<strong>Oncology</strong><br />

University <strong>of</strong> Jordan, P.O. Box 5321 Amman 11183, Jordan<br />

E-mail: salsukhun@yahoo.com<br />

Cancer is an increasing health problem, not only for industrialized countries but<br />

also for most other parts <strong>of</strong> the world.<br />

The large differences in cancer rates among countries, striking changes in these<br />

rates among migrating populations, and rapid changes over time within countries<br />

indicate that some aspect <strong>of</strong> lifestyle or environment is largely responsible for the<br />

common cancers in Western countries(1).<br />

Stomach cancer is one <strong>of</strong> the most common cancers worldwide. Almost twothirds<br />

<strong>of</strong> the cases occur in developing countries and 42% in China alone(1). The<br />

geographical distribution <strong>of</strong> stomach cancer is characterized by wide international<br />

variations; high-risk areas (Age adjusted incidence rate (ASR) in men, >20 per<br />

100,000) include East Asia (China, Japan), Eastern Europe, and parts <strong>of</strong> Central<br />

and South America. Incidence rates are low ( <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


Dietary fat has been hypothesized to be the key factor because national consumption<br />

is correlated with the international differences(6). However, detailed analyses<br />

in large prospective studies have not supported an important role <strong>of</strong> dietary<br />

fat. Instead, positive energy balance, reflected in early age at menarche and<br />

weight gain as an adult, is an important determinant <strong>of</strong> breast and colon cancers,<br />

consistent with numerous studies in animals(7&8). As a contributor to positive<br />

energy balance, and possibly by other mechanisms, physical inactivity has also<br />

been shown to be a risk factor for these diseases and in part accounts for the<br />

international differences (9&10). Although the percentage <strong>of</strong> calories from fat in<br />

the diet does not appear related to risk <strong>of</strong> colon cancer, greater risks have been<br />

seen with higher consumption <strong>of</strong> red meat, suggesting that factors other than fat<br />

per se are important. in prospective trials(11). On the other hand, a long standing<br />

belief in the protective role <strong>of</strong> fruits and vegetables was not confirmed. Taken<br />

together, the evidence supports a role for the triad <strong>of</strong> diet, body mass index and<br />

exercise in carcinogenesis.<br />

The Women Health Initiative (WHI) study is A randomized, controlled, primary<br />

prevention trial conducted at 40 US clinical centers from 1993 to 2005(12).<br />

A total <strong>of</strong> 48 835 postmenopausal women, aged 50 to 79 years, without prior breast<br />

cancer, were randomly assigned to the dietary modification intervention group (40%<br />

[n = 19 541]) or the comparison group (60% [n = 29 294]). The intervention was<br />

designed to promote dietary change with the goals <strong>of</strong> reducing intake <strong>of</strong> total fat<br />

to 20% <strong>of</strong> energy and increasing consumption <strong>of</strong> vegetables and fruit to at least 5<br />

servings daily and grains to at least 6 servings daily. Comparison group participants<br />

were not asked to make dietary changes. Surprisingly, that intervention did not<br />

result in a statistically significant reduction in invasive breast cancer risk over an<br />

8.1-year average follow-up period. However, the nonsignificant trends observed<br />

suggesting reduced risk associated with a low-fat dietary pattern indicated that<br />

longer, planned, nonintervention follow-up may yield a more definitive comparison.<br />

That same study failed to confirm the protective effect <strong>of</strong> normal levels <strong>of</strong> vitamin<br />

D against colorectal cancer suggested previously by the nurses’ health study(13).<br />

However, there were major limitations <strong>of</strong> that trial. Calcium and vitamin D was<br />

given as a combination. One <strong>of</strong> the limitations was just the fact that it was seven<br />

years, and that’s quite a short time in the development <strong>of</strong> colorectal cancer. It may<br />

be almost wishful thinking to believe that something like vitamin D or calcium<br />

could have an effect that quickly. Also, 69% <strong>of</strong> the women in the study were<br />

taking calcium supplements on their own, in both the placebo group and the active<br />

group, which causes huge misclassification. The controversial result from one <strong>of</strong><br />

the best well designed trials speaks for the complexity <strong>of</strong> conducting prospective<br />

trials to address the role <strong>of</strong> a single nutrient in cancer.<br />

The evidence does not only support a role for the triad in carcinogenesis, but also<br />

in mortality from cancer(14). In a prospectively studied population <strong>of</strong> more than<br />

900,000 US adults, increased body weight was associated with increased death<br />

rates for all cancers combined and for cancers at multiple specific sites, including<br />

esophagus, colon and rectum, liver, gallbladder, pancreas, and kidney; the same<br />

was true for death due to non-Hodgkin’s lymphoma and multiple myeloma. The<br />

authors estimated that current patterns <strong>of</strong> overweight and obesity in the United<br />

States could account for 14 percent <strong>of</strong> all deaths from cancer in men and 20 percent<br />

<strong>of</strong> those in women. Even more interesting were data suggesting midrange intake<br />

<strong>of</strong> most major energy sources was associated with best survival after treatment<br />

<strong>of</strong> breast cancer, and extremes were associated with less favorable outcomes(15).<br />

Recent data confirmed that vitamin E supplements, selenium supplements, or the 2<br />

<strong>of</strong> them taken together did not reduce the risk <strong>of</strong> developing prostate cancer (16).<br />

If anything, there was a small trend towards an increase in the number <strong>of</strong> prostate<br />

cancer cases for the men randomized to take vitamin E, and a small increase in<br />

the number <strong>of</strong> cases <strong>of</strong> type I I diabetes mellitus in men taking only selenium.<br />

In light <strong>of</strong> the above discussion, moderation <strong>of</strong> diet (quantity and quality) seems<br />

the most reasonable approach to adopt throughout life.<br />

References<br />

1. Parkin, D. M. et al. Global cancer statistics, 2002. CA Cancer J Clin 2005.<br />

2005 Mar-Apr;55(2):74-108.<br />

2. Kolonel LN et al. <strong>Association</strong> <strong>of</strong> diet and place <strong>of</strong> birth with stomach cancer<br />

incidence in Hawaii Japanese and Caucasians. Am. J. Clin. Nutr. 1981, 34:<br />

2478-2485.<br />

3. Tannenbaum A. The initiation and growth <strong>of</strong> tumours: Introduction, Effects <strong>of</strong><br />

undernutrition. Am J Cancer 1940, 35 : 335-350.<br />

4. Lavik PS, and Baumann CA. Further studies o the tumor promoting action <strong>of</strong><br />

fat. Cancer Res. 1943, 3: 749-756.<br />

5. Doll R, Peto R. Avoidable risks <strong>of</strong> cancer in the United States. J Natl Cancer<br />

Inst 1981;66:1196-1265.<br />

6. Overview and perspective in human nutrition. Willett WC. Asia Pac J Clin Nutr.<br />

2008;17 Suppl 1:1-4. Review.<br />

7. Hunter DJ, Willett WC. Diet, body size, and breast cancer. Epidemiol Rev<br />

1993;15:110-132.<br />

8. Chute CG, Willett WC, Colditz GA et al. A prospective study <strong>of</strong> body mass,<br />

height, and smoking on the risk <strong>of</strong> colorectal cancer in women. Cancer Causes<br />

Control 1991;2:117-124.<br />

9. Martinez ME, et al. Physical activity, body size, and colorectal cancer in women.<br />

Am J Epidemiol 1996;143:S73a.<br />

10. Giovannucci E, et al. Physical activity, obesity, and risk for colon cancer and<br />

adenoma in men. Ann Intern Med 1995;122:327-334.<br />

11. Giovannucci E, et al. Intake <strong>of</strong> Fat, Meat, and Fiber in Relation to Risk <strong>of</strong><br />

Colon Cancer in Men. Cancer Res. 1994, 54: 2390-2397.<br />

12. Prentice RL, et al. Low-Fat Dietary Pattern and Risk <strong>of</strong> Invasive Breast Cancer<br />

The Women's Health Initiative Randomized Controlled Dietary Modification Trial.<br />

JAMA. 2006;295:629-642.<br />

13. Wactawski-Wende J, et al, Calcium plus Vitamin D Supplementation and the<br />

Risk <strong>of</strong> Colorectal Cancer. N Engl J Med 354:684, February 16, 2006.<br />

14. Calle EE et al. Overweight, Obesity, and Mortality from Cancer in a<br />

Prospectively Studied Cohort <strong>of</strong> U.S. Adults, N Engl J Med 2003;348:1625-38.<br />

15. Goodwin P et al. Diet and Beast Cancer: Evidence That Extremes in Diet Are<br />

Associated with Poor Survival. J Clin Oncol 2003, 21:2500-2507.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 11


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

EPIDEMIOLOGY, PREVENTION AND MANAGEMENT GUIDELINES<br />

FOR BREAST CANCER IN ARAB COUNTRIES<br />

Nagi S. El Saghir, MD, FACP 1 , Omalkhair Abulkhair, MD 2<br />

(1) American University <strong>of</strong> Beirut <strong>Medical</strong> Center, Beirut, Lebanon<br />

(2) King Abdulaziz <strong>Medical</strong> City for National Guard, Riyadh, KSA<br />

Corresponding Author: Nagi S. El Saghir, MD, FACP<br />

Hematology-<strong>Oncology</strong>, American University <strong>of</strong> Beirut <strong>Medical</strong> Center<br />

Cairo Street, Ras Beirut, Beirut, Lebanon<br />

E-mail: nagi.saghir@aub.edu.lb<br />

Omalkhair Abulkhair, MD<br />

Department <strong>of</strong> <strong>Oncology</strong> (Mail code 1777), King Abdulaziz <strong>Medical</strong> City for<br />

National Guard, P.O. Box 22490, Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

E-mail: abulkhairo@ngha.med.sa<br />

Abstract<br />

Background: Breast cancer is the most common cancer among women in <strong>Arab</strong><br />

countries. Management varies according to human and financial resources, as<br />

well as available facilities.<br />

Materials and Methods: Literature review <strong>of</strong> publications on the epidemiology,<br />

prevention and treatment <strong>of</strong> breast cancer in <strong>Arab</strong> countries. Presentations and<br />

discussions summarized from various oncology meetings.<br />

Results: Breast cancer represents an average <strong>of</strong> one-third <strong>of</strong> female cancers in<br />

<strong>Arab</strong> countries. Incidence continues to rise and has reached up to 50/100.000<br />

women in Gulf countries and highest in Lebanon at 69/100000 women per year.<br />

Around 50% <strong>of</strong> cases are below the age <strong>of</strong> 50 years. Over 60% <strong>of</strong> cases present<br />

with advanced disease. However, number <strong>of</strong> advanced cases has started to decrease<br />

and more cases are being diagnosed at earlier stages because <strong>of</strong> recent awareness<br />

and early detection campaigns done in many <strong>Arab</strong> countries. Radiation therapy<br />

remains concentrated in major cities and large numbers <strong>of</strong> patients have to travel<br />

long distances to get adequate care. Multidisciplinary management is practiced<br />

only in major university hospitals and in the few available cancer centers.<br />

Conclusions: More efforts towards primary prevention are recommended.<br />

Secondary prevention includes awareness campaigns and early detection. Raising<br />

awareness, promoting attention to breast symptoms, breast self examination and<br />

clinical breast examination are recommended to reduce locally advanced breast<br />

cancer. In addition, screening mammography is recommended in countries with<br />

adequate resources. Although the starting age <strong>of</strong> screening mammography is<br />

controversial, we recommend to start at age 40 because 50% <strong>of</strong> cases <strong>of</strong> breast<br />

cancer in <strong>Arab</strong> countries occur in women below the age <strong>of</strong> 50. More group<br />

discussions and multidisciplinary management between individual physicians<br />

caring for women with breast cancer are needed.<br />

Introduction<br />

Cancer prevention and early detection are the most effective ways to control<br />

disease, alleviate sufferings, prolong survival, and eventually cure patients with<br />

cancer. Cancer prevention and early detection ultimately reduces the incidence <strong>of</strong><br />

cancer as well as morbidity and mortality from cancer. Early detection <strong>of</strong> breast<br />

cancer is considered as an important element <strong>of</strong> prevention <strong>of</strong> the disease. The<br />

improvement in breast cancer survival observed in industrialized nations in recent<br />

decades have been attributed to early detection by screening as well as to timely<br />

and effective treatment.1<br />

Consequently, breast cancer mortality which had been relatively unchanged from<br />

1930s through 1980s has dropped by 1.4% to 3.1% / year between 1990 and 2003<br />

in the US. 2 Cancer has become a national health priority according to the WHO<br />

in the Eastern Mediterranean Region. The general public, as well as healthcare<br />

pr<strong>of</strong>essionals, have noted that breast cancer has become one <strong>of</strong> the most important<br />

health problems for women in <strong>Arab</strong> countries. 3,4,5<br />

Approximately, 15% <strong>of</strong> all breast cancer cases can be attributed to familial and<br />

genetic influences.6 Most known risk factors for breast cancer can be linked<br />

to hazardous effects <strong>of</strong> prolonged exposure to estrogens7 such as early age at<br />

menarche, late age at menopause,7 small number <strong>of</strong> children and nulliparity, late<br />

age at first birth8 and little or no breast feeding9,10 which have all been associated<br />

with an increased risk <strong>of</strong> developing breast cancer. Although several retrospective<br />

studies have suggested that induced abortion is related to an increased risk <strong>of</strong><br />

this disease, this is not seen in prospective studies11 and its status as a breast<br />

cancer risk is unclear. Long term use <strong>of</strong> hormone replacement therapy has been<br />

associated with increased risk <strong>of</strong> developing breast cancer12 but apparently not<br />

use <strong>of</strong> oral contraceptives.12-15<br />

However, meta-analysis <strong>of</strong> all the data (Leslie Berstein, et al) ≥ ten years <strong>of</strong> use <strong>of</strong><br />

the relative risk 1.38 (these women were 1.38 times as likely to get breast cancer<br />

as those who had not used the pill). The primary concern is for women who start<br />

their period before age eleven. Those who also begin having regular periods much<br />

faster, so, prolonged use <strong>of</strong> estrogen containing pills is more likely to harm this<br />

subgroup.16 So far, there is no evidence that the use <strong>of</strong> mega-dose <strong>of</strong> hormone<br />

infertility treatment are hazardous to the breast.15,17 High breast density has<br />

been described as an important marker <strong>of</strong> breast cancer risk.18,19<br />

History <strong>of</strong> proliferative benign breast disease is also related to increased risk <strong>of</strong><br />

breast cancer20 especially those who underwent biopsies and those women in<br />

whom a typical hyperplasia was found in such biopsies.21 Therefore, in summary,<br />

breast cancer risk can be allocated to one <strong>of</strong> four groups: family history/genetic,<br />

reproductive/hormonal proliferative, benign breast disease with dysplasia, and<br />

mammographic density. Recently, it is believed that high-fat diet is not related per<br />

se to breast cancer risk22,23 but overall caloric intake and obesity in particular with<br />

weight-gain pattern, are related to increased breast cancer risk with differentiate<br />

effects between pre and post menopausal women.24,25<br />

Women with breast cancer may have advanced disease at diagnosis and some <strong>of</strong><br />

reasons for such presentations are summarized in Table 1.<br />

Table 1: Reasons For Presentation As Advanced Disease<br />

Lack <strong>of</strong> knowledge on breast cancer risk factors<br />

Misconception about breast cancer screening<br />

Fear <strong>of</strong> cancer<br />

Shyness and fear <strong>of</strong> social implications<br />

Beliefs and fatalistic attitudes<br />

Lack <strong>of</strong> health care facilities<br />

2. Disease Epidemiology<br />

Breast cancer is the most common cancer diagnosed in US women and the second<br />

leading cause <strong>of</strong> death <strong>of</strong> cancer in US women.1 The good news is that mortality<br />

from breast cancer has recently dropped slightly. This decrease has been attributed,<br />

in part, to mammographic screening.1, 2 Breast cancer has been reported as the<br />

most common cancer among women from many hospitals and medical centers in<br />

12 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


<strong>Arab</strong> countries. Hospital-based registries, several regional and national registries<br />

have recently been in place and reported their data. Researchers have reported data<br />

in either abstract form or meeting presentations and some reported in peer-reviewed<br />

local or international journals. Many national registries now publish their data<br />

as annual reports in a booklet format. In this manuscript, we review and update<br />

the available data on breast cancer epidemiology from <strong>Arab</strong> countries. 26, 27<br />

Breast cancer is the most frequent cancer in <strong>Arab</strong> women constituting 14% to 42%<br />

<strong>of</strong> all women cancers. Age-Adjusted Standardized incidence rates (ASR) were<br />

reported to vary from 9.5 to 50 cases per 100.000 women per year. Median age at<br />

presentation is 48-52 years and 50% <strong>of</strong> cases are below the age <strong>of</strong> 50; whereas only<br />

25% <strong>of</strong> cases in industrialized nations are below the age <strong>of</strong> 50 years, and 50% <strong>of</strong><br />

the cases are above the age <strong>of</strong> 63 years, 1, 2 thus it appears breast cancer in <strong>Arab</strong><br />

countries presents almost 10 years younger than that in USA and Europe26,27<br />

Age-adjusted standardized incidence rates (ASR) for breast cancer have increased<br />

in many <strong>Arab</strong> countries such as Lebanon (from 20 in 1996 to 46.7 in 1998 and<br />

even 69 in 2003), Jordan (ASR increased from 7.6/100.000 women in 1982 to<br />

32.8/100000 in 1997), Palestinians (ASR up by 93%), Egypt ASR up to 49.6.<br />

Reports from the Gulf Center for Cancer Registration (GCCR) which represented<br />

data from six Gulf countries: Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia, United <strong>Arab</strong> Emirates,<br />

Kingdom <strong>of</strong> Bahrain, Sultanate <strong>of</strong> Oman, State <strong>of</strong> Qatar and State <strong>of</strong> Kuwait,<br />

revealed that breast cancer is the most common in the GCC states between January<br />

1998 to December 2004. 6,882 breast cancer cases were reported from all GCC<br />

states accounted to 11.8% from all cancer and 22.7% from cancers among women.<br />

Bahrain reported the highest incidence.<br />

The ASR per 100,000 women was 46.4 followed by Kuwait 44.3, Qatar 35.5,<br />

UAE 19.2, Oman 14.4 and KSA 12.9. 28 Despite the low breast cancer incidence<br />

in the Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia (KSA), it is the most common cancer and it<br />

ranked number one in Saudi female population for the past 5 consecutive years<br />

(Saudi National Cancer Registry, 2000 – 2004). Data on female patients with<br />

invasive breast carcinoma reported from different regions in Saudi <strong>Arab</strong>ia revealed<br />

that most patients were between 40 – 50 years <strong>of</strong> age and were predominantly<br />

premenopausals. Although the rates are still below those in industrialized nations,<br />

they are rising and expected to reach the same levels. Changes may be due to<br />

Westernized life style changes including dietary habits, lack <strong>of</strong> exercise and<br />

urbanization, delay <strong>of</strong> ages <strong>of</strong> marriage and first pregnancy from the late teens<br />

and early twenties to the late twenties and even early thirties, respectively, as<br />

well as and decrease <strong>of</strong> the practice <strong>of</strong> breast feeding by <strong>Arab</strong> women. Advanced<br />

disease is commonly seen at presentation and in young women, diagnosis <strong>of</strong> breast<br />

cancer may be delayed because <strong>of</strong> decreased awareness as well as and low index<br />

<strong>of</strong> suspicion from their primary physicians. 29<br />

Young age at presentation has been shown to be a bad prognostic factor in two<br />

publications from Lebanon and Saudi <strong>Arab</strong>ia.30-31 (Figure-1)<br />

Figure-1 Young Age Confers a Worse Prognosis in Breast Cancer<br />

3. Risk Factors<br />

A number <strong>of</strong> breast cancer risk factors have been well established for many decades,<br />

notably those related to hormonal and reproductive exposures.7-8 For example,<br />

nulliparity late age at first childbirth, early age at menarche and use <strong>of</strong> hormone<br />

replacement therapy are all associated with increased risk.7-11 Additionally, the<br />

association <strong>of</strong> a family history <strong>of</strong> breast cancer with increased risk <strong>of</strong> the disease<br />

has long been established.32-33 The increased awareness <strong>of</strong> family history is<br />

enhanced partly by the establishment <strong>of</strong> the high risk, high penetrance BRCA1<br />

and BRCA2 germline mutations.34 Other risk factors pertaining to personal<br />

medical history has been established. These include diagnosis <strong>of</strong> atypical ductal<br />

hyperplasia and lobular carcinoma in situ.35 Prolonged exposure to birth control<br />

pills in premenopausal women, and especially hormone replacement therapy in<br />

post-menopausal women increase the risk <strong>of</strong> breast cancer. Other risk factors<br />

include radiation exposure, pollution and exposure to carcinogenic compounds<br />

such as pesticides and even smoking are suspected. Westernized dietary habits,<br />

increase consumption <strong>of</strong> animal fats and decreased fibers, fruits and vegetables,<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 13


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

as well as overweight and lack <strong>of</strong> exercise are known to increase breast cancer<br />

incidence among Asian immigrants to USA, Europe and Australia. Many <strong>of</strong> those<br />

risk factors are thought to apply to women in <strong>Arab</strong> countries. Only scattered studies<br />

on genetic mutations <strong>of</strong> BRCA-1, BRCA-2 and other oncogene mutations are<br />

available from <strong>Arab</strong> countries.36-37<br />

4. Presenting Signs and Symptoms<br />

The most common presentation in our countries is a breast lump or nodule,<br />

usually non-painful but may be associated with pain. Size <strong>of</strong> the lump depends<br />

on whether it was accidentally noted or whether the woman examines her breasts<br />

regularly. Other symptoms include changes in color or shape <strong>of</strong> the skin such as<br />

redness, ulceration and therapy <strong>of</strong> skin, nipple retraction or discharge <strong>of</strong> blood<br />

may be presenting complaints. A palpable mass in the axilla is not an uncommon<br />

presentation in cases <strong>of</strong> locally advanced breast cancer or without breast mass.<br />

Inflammatory breast cancer presents with a rapidly growing inflamed, thickened<br />

and red overlying breast skin. The data has shown that in many <strong>Arab</strong> countries<br />

that 50-80% <strong>of</strong> breast cancer is <strong>Arab</strong> countries present with advanced disease at<br />

presentation. More than 50% were Stage II and III while ductal carcinoma insitu<br />

represents <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


6. Management and Treatment Guidelines: Multidisciplinary Approach<br />

Breast cancer management must be composed <strong>of</strong> a multidisciplinary team that<br />

involves different specialty: breast surgeon/oncologist surgeon, medical oncologist,<br />

radiation oncologist, pathologist, psychologist, social worker, health educator and<br />

plastic surgeon. If a full team is not available, multi-disciplinary approach should<br />

be practiced by whoever is present. Even if the hospital where a breast cancer<br />

patient is treated has only a surgeon and a radiologist and pathologist, or a surgeon<br />

and radiologist and an oncologist, or a surgeon and a radiologist and a radiation<br />

oncologist, they should meet and discuss their patient’s management. Every<br />

effort should be made to have a pathologist available. Discussion and exchange<br />

information with physicians working at major cancer centers should be encouraged.<br />

If there is no multi-disciplinary clinic where the patient is seen by the full oncology<br />

team, every hospital that treats breast cancer should have a weekly Tumor Board<br />

conference where cases are discussed between available specialists.<br />

As a general role, the primary treatment is surgery. Partial mastectomy plus<br />

radiation therapy is equivalent to modified radical mastectomy for primary breast<br />

cancer. When partial mastectomy is performed, special attention should be made to<br />

obtain comfortably negative margins. Young women are at a higher risk <strong>of</strong> having<br />

local recurrences after long-term follow-up and should have continued long term<br />

follow-up. The treatment with mastectomy and reconstruction remains a viable<br />

option, particularly in younger women.<br />

Adjuvant therapy includes chemotherapy, targeted therapy with trastuzumab,<br />

and hormonal therapy. Guidelines are referenced below.42-43 Athracyclines and<br />

taxanes are the most commonly used drugs, either separately, or in sequence, or<br />

combination. Classical oral CMF remains a viable option. Attention to resources<br />

and health care planning in countries with limited resources are presented by<br />

Breast Health Global Initiative (BHGI) .44<br />

Locally advanced breast cancer is treated with neoadjuvant therapy, preferably<br />

and more clearly called pre-operative therapy. Pre-operative therapy is usually<br />

anthracycline-based. 45 Taxanes improve breast conservation rates and increase<br />

complete pathological remissions46 Patients with less residual disease tend to<br />

have better survival.47 Higher rates <strong>of</strong> clinical and pathological remissions may<br />

be obtained with chemotherapy and targeted therapy in the presence <strong>of</strong> HER2<br />

positive tumors. Hormonal preoperative therapy gives good clinical responses<br />

produces but rarely produces complete pathological remissions and is useful in<br />

older postmenopausal patients. Preoperative therapy requires adequate radiological<br />

and pathological evaluations to be available.48<br />

Guidelines for the treatment <strong>of</strong> metastatic disease are referenced below. In general,<br />

hormonal therapy is indicated for hormone-receptor positive metastatic disease<br />

with bone and s<strong>of</strong>t tissue metastases. Chemotherapy is used in patients with<br />

visceral metastases such as in the liver and lung. Chemotherapy is generally<br />

used when patients have aggressive and rapidly progressive disease particularly<br />

in young patients. Ovarian ablation (surgical, or by radiation therapy, or more the<br />

more costly ovarian suppression by LHRH analogs is preferred in premenopausal<br />

women. Tamoxifen with ovarian ablation is better than tamoxifen alone. Tamoxifen<br />

is effective anti-estrogenic therapy also in post-menopausal patients. Aromatase<br />

Inhibitors, particularly letrozole has been shown to be more effective than<br />

tamoxifen. Attention to resources and availability <strong>of</strong> therapy is important in<br />

countries with limited resources and has started to gain attention in major medical<br />

conferences and journals as referenced below.48<br />

Targeted therapy with anti-HER2 agent trastuzumab has become an essential<br />

component <strong>of</strong> the treatment <strong>of</strong> CerbB2 (HER2/neu) positive disease. Trastuzumab<br />

may be combined with hormonal therapy (TanDEM trial) or chemotherapy<br />

particularly docetaxel, paclitaxel and vinorelbine. More recently, lapatinib, a<br />

dual anti-Her2 and tyrosine kinase inhibitor has been reported to be beneficial in<br />

trastuzumab-resistant patients and also upfront when effectively combined with<br />

hormonal therapy in combination with letrozole and lapatinib in breast cancer<br />

(Abstract #81/ 31st Annual San Antonio Breast Cancer Symposium, 2008).<br />

Modern treatment <strong>of</strong> breast cancer should include research and clinical trials to<br />

find better answers for many questions. Modern chemotherapy, targeted therapy,<br />

and new hormonal therapy require more research and participation in international<br />

phase III trials. The medical community in the <strong>Arab</strong> World is urged to establish<br />

nationwide and hospital computerized databases and publish its findings and<br />

its research in peer-reviewed scientific journals. Multidisciplinary approach to<br />

cancer management is currently practiced only in few major medical centers and<br />

should become standard practice in all hospitals that treat breast cancer. More<br />

cooperation between various specialists to improve care <strong>of</strong> breast cancer patients<br />

is direly-needed. A few cancer centers exist in <strong>Arab</strong> countries and many more are<br />

needed. Encouragement and funding <strong>of</strong> cancer research should become a priority<br />

in <strong>Arab</strong> countries. Readers are referred to Breast Health Global Intitiative BHGI<br />

resource-oriented guidelines published in the October 2008 issue <strong>of</strong> Cancer.48<br />

7. Prevention Through Risk Factor Modification<br />

It is clear that modern therapy for breast cancer has become more effective and<br />

more expensive. Prevention and early detection may save major parts <strong>of</strong> costs<br />

<strong>of</strong> adjuvant and metastatic disease therapy. Several strategies are available for<br />

reducing breast cancer risk in countries with lower resources, but few <strong>of</strong> them<br />

have completed rigorous testing in clinical trials.28 Strategies to increase the<br />

prevalence and length <strong>of</strong> lactation may reduce risk for breast cancer in mothers in<br />

addition to providing nutrition benefits for infants and small children. Increased<br />

adiposity, a sedentary lifestyle and moderate to high levels <strong>of</strong> alcohol use are<br />

associated with increased risk <strong>of</strong> breast cancer. The evidence <strong>of</strong> a role for specific<br />

dietary components is less clear. For individual women, counselling should include<br />

increasing physical activity and balancing energy such that weight remains stable<br />

over a lifetime and, preferably, with the body mass index remaining


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

Table 3: Prevention Through Risk Factor Modification<br />

Risk Factors Suggested Modification<br />

Breast feeding To prolong lactation ≥ 1 ½ years<br />

at least as many studies proved its<br />

beneficial protection.<br />

Lifestyle and diet Increase mobilization, avoid weigh<br />

gain specially after menopause,<br />

regular exercise.<br />

Smoking, alcohol Avoid smoking and alcohol.<br />

Hormone Replacement Therapy<br />

(HRT)<br />

Should not be given routinely to<br />

postmenopausal women. Should<br />

not be dispensed from pharmacies<br />

without prescription.<br />

8. Early Detection: Experience in <strong>Arab</strong> Countries<br />

In the majority <strong>of</strong> <strong>Arab</strong> countries, screening is not standard <strong>of</strong> care. However, few<br />

promising screening initiatives emerged recently in some countries. The Cairo<br />

Breast Cancer Screening Trial was reported in 2005. It included training nurses<br />

and social workers to provide lectures and demonstarations, breast examinations,<br />

and referring women with suspicious findings to a nearby Italian Hospital in<br />

Cairo. More early breast cancer cases and less mastectomies were reported by<br />

the investigators.39 Another major effort is being conducted with the Ministry<br />

<strong>of</strong> Health.<br />

8.1. Early Detection: Experience in Saudi <strong>Arab</strong>ia<br />

In Saudi <strong>Arab</strong>ia, several studies were done evaluating the perceptions <strong>of</strong> women<br />

regarding breast cancer screening. In addition to physician barriers to breast<br />

cancer screening, studies identified the following factors: lack <strong>of</strong> knowledge about<br />

mammography as it is the standard screening test, breast cancer risk factors and<br />

lack <strong>of</strong> knowledge <strong>of</strong> practice BSE.51, 52<br />

The study that targeted primary health care physician concluded that the main<br />

barriers to breast cancer screening were: the unavailability <strong>of</strong> a national screening<br />

program, lack <strong>of</strong> women cooperation and compliance, lack <strong>of</strong> allocated time by<br />

physicians, and lack <strong>of</strong> knowledge regarding screening recommendations. 53<br />

Recently, two screening programs were developed; one in Al-Qaseem region where<br />

investigators developed a program for early detection <strong>of</strong> breast cancer that included<br />

staffing by trained nurses, pathologists and radiologist and target 75,000 female<br />

ages 35-60 in the region. The program has five permanent screening centers and<br />

a mobile one. So far, they completed three years since they started but no <strong>of</strong>ficial<br />

results, for their findings are available yet (Alhabdan, personal communication).<br />

The second screening center is the Abdul-Lateef Center for Screening in the capital<br />

city <strong>of</strong> Riyadh, which opened in October 2007. The center is equipped with a digital<br />

mammogram machine, an ultrasound and two radiologists for double reading <strong>of</strong><br />

mammographies, in addition to general practitioner and health educator who will<br />

interview the women and provide them with the screening questionnaires. For<br />

women with average risk factors using Gail Model questionnaires, a mammogram<br />

will be done based on BIRADS System (taken from http:/www.imaginis.com/<br />

breasthealth/acrbi.asp). After a screening mammography is generated, the women<br />

are contacted. Those who need further evaluation are sent to King Abdulaziz<br />

<strong>Medical</strong> City or King Fahad <strong>Medical</strong> City in Riyadh.. So far, over 1,800 females<br />

were screened and 22 women were confirmed to have malignancies. A few <strong>of</strong><br />

those malignancies were ductal carcinoma insitu (DCIS) and Stage 1 disease.<br />

Investigators are encouraged and women are re-assured that we see early breast<br />

cancer rather than the usual advanced disease. Challenges facing the investigators<br />

include refusal <strong>of</strong> many women to do further investigations, and even some<br />

women refused to have treatment. Those issues highlight the importance <strong>of</strong> breast<br />

health education for the general population. Results <strong>of</strong> this program are accepted<br />

presentation in ASCO 2009.<br />

8.2 Experience from Lebanon<br />

In addition to general public awareness campaigns, there is a systemic campaign<br />

that started in 2003, in collaboration with the Ministry <strong>of</strong> Health, Lebanese Society<br />

<strong>of</strong> <strong>Medical</strong> <strong>Oncology</strong>, Non Governmental Organization Faire Face (Facing Cancer),<br />

and pharmaceutical company H<strong>of</strong>fman-La Roche. Every October, the campaign<br />

runs billboard ads, TV ads, intense TV and newspaper and magazine interviews<br />

about breast cancer screening and education. The campaign recently evolved into<br />

getting <strong>of</strong>fers for low-cost mammography <strong>of</strong>fered once a year for women over<br />

40. The Campaign and Ministry <strong>of</strong> Health <strong>of</strong>fer classes and guidelines for quality<br />

mammography for participating centers. In parallel to the campaigns, there has<br />

been a noticeable increase in mammography-detected cancers, small tumors, and<br />

less total mastectomies being performed at major institutions.42 A National Breast<br />

Cancer Task Force has evolved to plan further actions.<br />

8.3. Breast Cancer Screening Recommendations<br />

The recommendation <strong>of</strong> breast cancer early detection/screening in average risk<br />

and high risk women are depicted in Tables 4 and 5. These guidelines should be<br />

followed in the <strong>Arab</strong> countries until future relevant evidences prove the need to<br />

modify these guidelines.<br />

Table 4: American Cancer Society (ACS) and National Comprehensive Cancer<br />

Network (NCCN) Recommendations for the Early Detection <strong>of</strong> Breast Cancer<br />

in Average-Risk Asymptomatic People<br />

Population Test or<br />

Procedure<br />

Frequency<br />

Women, aged Breast self- Beginning in their early 20s, women<br />

≥20 years but examination should be told about the benefits and<br />

40 years<br />

Clinical breast<br />

examination<br />

(CBE)<br />

Mammography Not recommended<br />

Breast self- Optional<br />

examination<br />

(BSE)<br />

Clinical breast Annually<br />

examination<br />

(CBE)<br />

Mammography** Annually<br />

Recommended that CBE be part <strong>of</strong> a<br />

periodic health examination, preferably<br />

at least every 3 years.<br />

16 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


* The ACS no longer recommends monthly (BSE) and women should be<br />

informed about the potential benefits, limitations and harms (the possibility <strong>of</strong><br />

References<br />

false positive result). Also, women should receive instructions in the technique 1. Ries Lag, Melbert D, Krapcho M, et al. SEER Cancer Statistics Review. 1975and/or<br />

have technique reviewed periodically.<br />

2004 National Cancer Institute. Bethesda MD. http//seer.cancer.govt/cst/1975-2004<br />

** There is no specific upper age at which mammography screening should Based on Nov. 2006 SEER data submission. Posted to the SEER website. 2007<br />

be discontinued, decision to stop annual mammography screening should be Weif, Miglioretti DI, Connelly MT, et al. Changes in women use <strong>of</strong> hormones after<br />

individualized based on potential benefits and risk <strong>of</strong> screening in the context <strong>of</strong> the women’s health initiative estrogen and progesterone trial by race, education<br />

overall health status and estimated longevity.<br />

and income. J Natl Cancer Inst Monogr 2005 (35): 106-112<br />

2. Jemal A, Ward E, Thun MJ. Recent trends in breast cancer incidence rates<br />

Table 5: American Cancer Society (ACS) and National Comprehensive Cancer by age and tumor characteristics among US women. Breast Cancer Res. May<br />

Network (NCCN) Recommendations for the Early Detection <strong>of</strong> Breast Cancer 3, 2007, 9 (3): R28<br />

in High-Risk Asymptomatic People<br />

3. The World Health Organization Fight <strong>Against</strong> Cancer: Strategies that prevent,<br />

Population Test or Procedure Frequency<br />

cure and care. 2007. www.who.int/cancer/en<br />

Prior thoracic radiation<br />

therapy (RT) for age<br />

Breast self-examination<br />

(BSE)*<br />

Periodic monthly<br />

4. Women’s News 10/05/06 U.S. Groups Join Saudi Breast Cancer Effort<br />

5. Laura Bush in Saudi <strong>Arab</strong>ia to Promote Breast Cancer Awareness. Wikinews’<br />


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

2008. 148:337-3347<br />

20. Wang J, Constantine JP, et al. N. Lower-category benign breast disease and<br />

the risk <strong>of</strong> invasive breast cancer. J Natl Cancer Inst 2004; 96:616-20<br />

21. Vogel VG. Atypia in the assessment <strong>of</strong> cancer risk: Implication for management<br />

design cytophatol 2004;30:151-7<br />

22. Cho E, Spiegelman D, Hunter DJ, et al. Premenopausal fat intake and risk <strong>of</strong><br />

breast cancer. J Natl Cancer Inst 2003;95:1079-85<br />

23. Velie E, Kuldorff M, et al. Dietary fat subtypes and breast cancer in<br />

postmenopausal women: A prospective cohort study. J Natl Cancer Inst 2000,<br />

92:833-9<br />

24. Key T, Appleby PN, et al. Body mass index, serum sex hormones and breast<br />

cancer risk in postmenopausal women. J Natl Inst 2003; 95;1218-26<br />

25. Harvie M, Howell A, et al. <strong>Association</strong> <strong>of</strong> gain and loss <strong>of</strong> weight before and<br />

after menopause with risk <strong>of</strong> post menopausal <strong>of</strong> breast cancer in Iowa Women’s<br />

Health Study. Cancer Epidemiol Biomarkers 2005; 14:656-61 26 NCR 2002,<br />

2003, 2004<br />

27. El Saghir NS, Khalil MK, et al. Trends in epidemiology and management <strong>of</strong><br />

breast cancer in developing <strong>Arab</strong> countries: A literature and registry analysis.<br />

Int J Surg 2002 Aug; 5 (4):225-33<br />

28. Gulf Center for Cancer Registry (GCCR). 2004<br />

29. M. Lamyian, A. Hydrania, et. Al. Barriers to and factors facilitating breast<br />

cancer screening among Iranian Women : A qualitative study. Health <strong>Journal</strong> Vol<br />

13 No 5: Sept. – Oct. 2007<br />

30. Nasser Elkum, Said Dermime, et al. Being 40 or younger is an independent risk<br />

factor for relapse in operable breast cancer patients: The Saudi <strong>Arab</strong>ia experience.<br />

BMC Cancer 2007, 7:222 doi:10.1186/1471-2407-7-222<br />

31. Nagi El Saghir, Muhieddine Seoud, et al. Effects <strong>of</strong> young age at presentation on<br />

survival in breast cancer. BMC Cancer 2006, 6:194:doi:10.1186/1471-2607-6-194<br />

32. Thompson WD. Genetic epidemiology <strong>of</strong> breast cancer. Cancer 1994;74:279-<br />

287<br />

33. Collaborative Group on Hormonal Factors in Breast Cancer. Familial breast<br />

cancer: collaborative reanalysis <strong>of</strong> individual data from 52 epidemiological<br />

studies including 58,209 women with breast cancer and 101,986 women without<br />

the disease. Lancet 2001; 358(9084):1047-1059.<br />

34. Ford D, Easton DF, Stratton M, Narod S, Goldgar D, Devilee P, Bishop DT,<br />

Weber B, Lenoir G, Chang-Claude J, Sobol H, Teare MD, Stuewing J, Arason<br />

A, Scherneck S, Peto J, Rebbeck TR, Tonin P, Neuhausen S, Barkardottir R,<br />

Eyfjord J, Lynch H, Ponder BA, Gayther SA, Zeladad-Hedman M et al. Genetic<br />

heterogeneity and penetrance analysis <strong>of</strong> BRCA1 and BRCA 2 genes in breast<br />

cancer families. The Breast Cancer Linkage Consortium. American <strong>Journal</strong> <strong>of</strong><br />

Human Genetics 1998;62:676-689.<br />

35. Page DL, Kidd Jr TE, Dupont WD, Simpson JF, Rogers LW. Lobular neoplasia<br />

<strong>of</strong> the breast : higher risk for subsequent invasive cancer predicted by more<br />

extensive disease. Human Pathology 1991;22(12):1232-1239.<br />

36. El Harith E, Abdel-Hadi MS, et al. BRCA1 and BRCA2 mutations in breast<br />

cancer patients from Saudi <strong>Arab</strong>ia. Saudi Med J 2002;23:700-4<br />

37. Bedwani R, Abdelfattah M, et al. Pr<strong>of</strong>ile <strong>of</strong> familial breast cancer in Alexandria,<br />

Egypt. Anticancer Res 2001;21:3011-4.<br />

38. Pestalozzi B, Catiliogne M; ESMO Guidelines Working Group. Primary breast<br />

cancer: ESMO clinical recommendations for diagnosis, treatment and follow-up.<br />

Ann Oncol. 2008 May;19 Suppl 2:ii7-10.<br />

39. Boulos S, Gadallah M, Neguib S, Essam E, Youssef A, Costa A, Mittra I, Miller<br />

AB. Breast screening in the emerging world: high prevalence <strong>of</strong> breast cancer in<br />

Cairo. Breast. 2005 Oct;14(5):340-6<br />

40. Abulkhair O. Abstract at ASCO Breast Cancer Symposium. A. Omalkhair et<br />

al: 2007 Breast Ca Symposium<br />

41. Goldhirsch A, Wood WC, Gelber RD, Coates AS, Thurlimann B, Senn HJ;<br />

10th St. Gallen conference. Progress and promise: highlights <strong>of</strong> the international<br />

expert consensus on the primary therapy <strong>of</strong> early breast cancer 2007. Ann Oncol.<br />

2007 Jul;18(7):1133-44. Review. Erratum in: Ann Oncol. 2007 Nov;18(11):1917<br />

42. El Saghir N. Abstract at ASCO Breast Cancer Symposium. El Saghir N et al:<br />

2007 Breast Ca Symposium<br />

43. NCCN Clinical Practice Guidelines in <strong>Oncology</strong><br />

http://www.nccn.org/pr<strong>of</strong>essionals/physicians_gls/PDF/breast.pdf; accessed 20<br />

September 2008<br />

44. Anderson BO, Yip CH, Smith RA, et al. Guideline implementation for breast<br />

healtcare in low and middle income countries: overview <strong>of</strong> the breast health global<br />

initiative summit 2007. Cancer. 2008;113(8 suppl):2221-2242<br />

45. Mauri et al. JNCI 2005 consensus article: Mauri D, Pavlidis N, Ionnidis JP.<br />

Neoadjuvant versus adjuvant systemic treatment in breast cancer : A meta-analysis.<br />

J Natl Cancer Inst. 2005;97:188-194.<br />

46. Wolmark NCI consensus conference update B-27, NCI 2007 website reference.<br />

47. P. F. Escobar, R. Patrick, et al. Clinical predictors with residual breast<br />

disease after primary induction chemotherapy. JCO;2004 ASCO Annual meeting<br />

proceeding. Vol 2, No 145 (July 15 supplement) 2004;688<br />

48. El Saghir NS, Eniu A, Carlson RW, Aziz Z, Vorobi<strong>of</strong> D, Hortobagyi GN. Locally<br />

advanced breast cancer: treatment guidelines with particular attention to low- and<br />

middle-income countries. Cancer. 2008;113(8 suppl):2314-2323<br />

49. Patel AV, Callel EE, et al. Recreational physical activity and risk <strong>of</strong> post<br />

menopausal breast cancer in a large cohort <strong>of</strong> US women. Cancer causes control<br />

2003;14:519-29<br />

50. Collaborative Group on Hormonal Factors in Breast Cancer. Alcohol,<br />

tobacco and breast cancer – collaborative reanalysis <strong>of</strong> individual data from 53<br />

epidemilogical studies including 58,515 women with breast cancer and 95,067<br />

women without the disease. BRJ Cancer 2002;87:1234-45<br />

51. Awahf Alam. Lack <strong>of</strong> knowledge <strong>of</strong> breast cancer risk factor and protection:<br />

BSE Knowledge. Annals <strong>of</strong> Saudi Medicine. 2006: Vol 26(4) Jul-Aug 2006;2727<br />

52. Khadiga F. Dandash, Abdulrahman Al-Mohaimeed. Knowledge, attributes and<br />

practice surrounding breast cancer and screening in female teachers <strong>of</strong> Buraidah,<br />

Saudi <strong>Arab</strong>ia. International <strong>Journal</strong> <strong>of</strong> Health Science. Vol 1, Jan 2007<br />

53. Layla A. Al-Alaboud, et al. The barriers <strong>of</strong> breast cancer screening program<br />

among PHHC female physicians. Middle East <strong>Journal</strong> <strong>of</strong> Family Medicine. Sept.<br />

2006; vol 6 Issue 5<br />

18 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


CANCER MAGNITUDE IN SHAM COUNTRIES<br />

Khatib S. 1 , Al-Tarawneh M. 2<br />

(1) King Hussein Institute for Biotechnology and Cancer<br />

(2) Ministry <strong>of</strong> Health, Jordan<br />

Corresponding Author: Dr. Sami Khatib<br />

Senior Assistant Director General, King Hussein Institute for Biotechnology and<br />

Cancer (KHIBC), Amman, Jordan<br />

E-mail: skhatib@khibc.jo<br />

Objective<br />

This study was designed to present and highlight some epidemiological<br />

characteristics <strong>of</strong> cancer in Sham countries<br />

Methodology<br />

Epidemiological data on cancer in Sham countries as in many <strong>Arab</strong> countries<br />

are limited. In this study we searched Pub Med, Medline, WHO, Globocan and<br />

MECC publications, national cancer registries and abstracts where it is available.<br />

We analyzed available data on some epidemiological characteristics <strong>of</strong> cancer in<br />

Sham countries and compared it to other <strong>Arab</strong> countries (KSA, Oman, Kuwait,<br />

Egypt, Tunisia).<br />

Results<br />

It was found that cancer <strong>of</strong> all sites is more predominant in females with a male<br />

to female ratio less than 1 in all Sham countries except in Syria where it is the<br />

reverse. The median age at diagnosis <strong>of</strong> cancer in all Sham countries is within the<br />

range <strong>of</strong> 52-56 years. The age standardized rates for males (ASR per 100,000 male<br />

populations) in Sham countries are as follow (Jordan, Lebanon, Syria, Palestinewest<br />

bank, 113, 179, 166, 128, respectively), it is higher than that in KSA, Oman,<br />

but less than that in Egypt. The same was found for the ASR in females. The five<br />

leading cancers among men in Sham Countries are: Jordan; colo-rectal (11.4%)<br />

leukemia (11.2%) lung (10.7%) urinary bladder (8.1%) prostate (7.6%) Lebanon<br />

as follow: Prostate (16.5%) urinary bladder (16%) lung (15%) colorectal (7.9%)<br />

and lymphoma 4.8%. In Palestine-West bank as follow: lung (12.4%) lymphoma<br />

(9.6%) colorectal (9.2%) prostate (9.1%) and urinary bladder (8.4%). In Syria,<br />

the five leading cancers among men as follow Lung (11.8%), urinary bladder<br />

(9.8%), Colorectal (9.6%), prostate (9.3%), N.H. Lymphoma (8.1%). Whereas<br />

among women the 1st rank cancer in all Sham countries is the breast cancer with<br />

relative frequency as follow (Jordan, Lebanon, Palestine, 34.8%, 40.2%, 27.2%,<br />

respectively) except in Syria where the uterus cancer is the 1st (32.6 %). The<br />

2nd is colorectal (Jordan, Lebanon, Palestine, Syria, 9%, 7.6%, 27.6%, 9.4%,<br />

respectively). ASR for colo-rectal cancer was found to be similar for all Sham<br />

countries and approximately the same for men and women (7 for men and 6 for<br />

women, also it’s similar to ASR in KSA, Oman Egypt and Tunisia (except Lebanon<br />

15.5 for men, 14.2 for women per 100,000 populations its similar to that found in<br />

Kuwait. ASR for lung cancer is 4 times higher in males than females in all Sham<br />

countries. The ASR for bladder cancer is higher in males than females in all Sham<br />

countries with a range from 1.3-4.8). The other cancers (stomach, lymphoma,<br />

leukemia); the ASR is higher in males than females. The ASR for prostate cancer<br />

was found to be similar in all Sham countries and the <strong>Arab</strong> countries included in<br />

the analyzes. The highest ASR for breast cancer was found in Lebanese females<br />

(69.2/100.000) meanwhile the lowest rate was found in Syria (31.2/100.000).<br />

Conclusions and recommendations<br />

Cancer incidence is on the rise in all Sham countries as in other <strong>Arab</strong> countries<br />

where incidence rates for both men and women is increasing. Comprehensive<br />

cancer control programs are highly needed. Inter-countries cooperation and<br />

collaboration is recommended.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 19


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

THE ROLE OF MRI ON CANCER DETECTION AND MANAGEMENT:<br />

THE SAUDI ARABIAN EXPERIENCE<br />

Aldabbagh A. 1<br />

(1) King Abdulaziz University Hospital, Jeddah, KSA<br />

Corresponding Author: Pr<strong>of</strong>. Dr. Asma Aldabbagh<br />

Pr<strong>of</strong>essor and Consultant in Radiology, Radiology Department<br />

Faculty <strong>of</strong> Medicine & Allied Sciences, King Abdulaziz University Hospital<br />

P.O. Box 80215, Jeddah 21589, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

Introduction<br />

MRI is increasingly being used as a problem solving tool; Examination timing,<br />

indications, and technique are critical to its success. It is particularly useful in<br />

mammographically-dense breasts, patients presenting with abnormal axillary<br />

nodes, the post-operated breast and to monitor neoadjuvant therapy response. It<br />

has also been recommended for screening high risk patients.<br />

Disease Epidemiology<br />

Breast Cancer in Saudi <strong>Arab</strong>ia is the most common cancer in women. National<br />

screening is as yet not implemented. Most women usually come because <strong>of</strong><br />

symptoms. Many <strong>of</strong> the cancers discovered are at an advanced stage.<br />

Screening Methods Overview<br />

Although mammography remains the primary imaging modality for the detection<br />

and evaluation <strong>of</strong> breast cancer, additional imaging is usually required to further<br />

stage the disease so that preoperative or other management strategies are planned.<br />

MRI is superior to other modalities in the detection <strong>of</strong> occult disease in the same or<br />

contralateral breast. MRI is also superior than other breast imaging modalities in the<br />

evaluation <strong>of</strong> the post operative breasts, monitoring neoadjuvant chemotherapeutic<br />

response and in the assessment <strong>of</strong> high risk patients.<br />

In our hospital, 1226 patients had breast MRI examinations. 406 patients were<br />

thought to have malignant lesions based on MRI findings. 298 cases were<br />

histopathologically proven malignancies. Of the proven malignant cases, 28<br />

patients gave a family history <strong>of</strong> breast cancer. 71 patients continued their<br />

investigations elsewhere and histopathology results were not available. 35 cases<br />

were false positive. Only 2 cases were false negative. Breast cancer recurrence<br />

was diagnosed by MRI in 42 patients, 8 <strong>of</strong> those were histopathologically negative<br />

for malignancy. Only a few patients were monitored by MRI during the course<br />

<strong>of</strong> their Neoadjuvant therapy.<br />

Outcome and Recommendations<br />

• Contrast-enhanced MRI has a high sensitivity and a high negative predictive<br />

value for the detection <strong>of</strong> invasive breast cancer.<br />

• It is superior to mammography and sonography for the detection <strong>of</strong><br />

recurrence.<br />

• Breast MRI should be interpreted in conjunction with other breast imaging<br />

modalities.<br />

• Patients' history with dates <strong>of</strong> previous treatments should be given.<br />

• As with mammography, previous MRI studies availability is helpful.<br />

• It is also logical that lesions found only on MRI are biopsied under MRI<br />

guidance, a situation which is difficult to undertake in most hospitals if a<br />

dedicated breast MRI unit is not available.<br />

• MRI should be used to screen high risk patients.<br />

• MRI has made a significant impact on breast cancer detection and management<br />

in our practice.<br />

References<br />

1. Christiane K. Kuhl, Current Status <strong>of</strong> Breast MR Imaging. RSNA, 2007.<br />

2. Laura Liberman et al, Breast Lesions Detected on MR Imaging: Features and<br />

Positive Predictive Value, AJR 2002; 179:171-178.<br />

3. Elizabeth Morris, Laura Liberman, Breast MRI Diagnosis and Intervention.<br />

4. Constance D. et al. Cancer Yield <strong>of</strong> Mammography, MR, and US in High-Risk<br />

Women: Prospective Multi-Institution Breast Cancer Screening Study. Radiology<br />

2007;244:381-388.<br />

5. Ansgar Malich et al, Potential MRI Interpretation Model: Differentiation <strong>of</strong><br />

Benign from Malignant Breast Masses. AJR 2005; 185:964–970<br />

20 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


ONCOLOGIST PERSPECTIVE ON TOBACCO CONTROL:<br />

HISTORICAL VIEW AND PRACTICAL GUIDELINES<br />

Mushabbab Asiri MD FFR RCSI FRCPC 1 , Abdullah Al Amro MD FRCPC 3 ,<br />

Sulaiman Al Sobai 3 and Abdul Rahman Jazieh, MD, MPH 4<br />

(1) Department <strong>of</strong> <strong>Oncology</strong> Riyadh Military Hospital and Prince Sultan<br />

Haematology and <strong>Oncology</strong> Center King Fahad <strong>Medical</strong> City Riyadh, Saudi<br />

<strong>Arab</strong>ia, Saudi Cancer Society and Prince Sultan Haematology and <strong>Oncology</strong><br />

Center King Fahad <strong>Medical</strong> City Riyadh, Saudi <strong>Arab</strong>ia,<br />

(3) Saudi Anti Smoking Organization, Riyadh, Saudi <strong>Arab</strong>ia<br />

(4) Department <strong>of</strong> <strong>Oncology</strong>, King Abdulaziz <strong>Medical</strong> City, Riyadh, Saudi <strong>Arab</strong>ia<br />

Corresponding Author: Mushabbab Asiri<br />

Director Department <strong>of</strong> <strong>Oncology</strong> Riyadh Military Hospital, PO BOX 295868,<br />

Riyadh 11351, Saudi <strong>Arab</strong>ia<br />

E mail: mushabbabasiri@gmail.com / maasiri@rmh.med.sa<br />

Abstract<br />

Background: Tobacco use is a major risk factor for multiple health problems<br />

including many types <strong>of</strong> cancer. It is estimated that about third <strong>of</strong> the cancer cases<br />

are attributed to tobacco exposure.<br />

Methods: Review <strong>of</strong> the literature about tobacco use and its associated risk in<br />

cancer development was conducted and summarized in addition to the local<br />

experience in tobacco control initiatives and program.<br />

Results: Tobacco is a rising healthcare concern in the region that will lead to<br />

significant increase in cancer cases. Various smoking cessation program and<br />

interventions are identified in addition to potential roles <strong>of</strong> physician in combating<br />

these deadly habits. Recommendations to the practicing health provider in the<br />

region were suggested. Various recommendations about physician roles in this<br />

issue were also reviewed.<br />

Conclusion: In spite <strong>of</strong> its known risk to health, tobacco use remains a growing<br />

health concerns. Individual health care providers, organizations, and government<br />

need to carry well orchestrated efforts to minimize tobacco associated risks.<br />

Introduction and History<br />

Smoking is a practice where the substance such tobacco is burned and its smoke<br />

inhaled, the composition releases the active ingredients such as nicotine making<br />

it available for absorption through the lungs.<br />

Smoking evolved in association with a ritual ceremony 7000 years ago originated<br />

in Peruvian and Ecuadorian Andes and was found in several ancient civilizations<br />

such as Indian, Chinese, Babylonians, Greece, Shamans, Maya and Aztec tribes<br />

in South America (1)<br />

Hashishah (cannabis) smoking was known in the Middle East for several hundred<br />

years before the arrival <strong>of</strong> tobacco.<br />

Using water pipe called (hookah) in Urdu language was common practice in the<br />

Middle East and the water pipe (Shishah, Argelah) was a major part <strong>of</strong> wedding<br />

gifts in several parts <strong>of</strong> Middle East especially Iran and Turkey. This habit has<br />

spread to other areas during Othman empire.(2,3)<br />

The European adopted the smoking habits subsequent to colonization <strong>of</strong> the<br />

Americas in the 16th century. Jean Nicot, a French diplomat, brought tobacco<br />

plant from Portugal to France and introduce it to the high class family <strong>of</strong> France<br />

whom call it Nicotiana plant after his name in 1560. From his name, the word<br />

nicotine was derived, tobacco smoking spread from France to England and to<br />

the rest <strong>of</strong> Europe.(4)<br />

Inhalation <strong>of</strong> substances during the inspiration will deliver the active substances in<br />

the smoked materials into the circulation very effectively because the lung contains<br />

millions <strong>of</strong> alveoli and this route <strong>of</strong> drug administration is commonly used in the<br />

treatment <strong>of</strong> bronchial asthma and in anesthesia that it is fast and effective but<br />

unfortunately this route had been abused by recreation drug abusers.<br />

Tobacco is plant that its leaves processed for the production <strong>of</strong> smoked tobacco that<br />

is used for chewing or sniffing <strong>of</strong> cigarettes, cigar, pipe and hookah (Sheeshah).<br />

Modern cigarette made <strong>of</strong> blended shredded tobacco leaf and tobacco dust processed<br />

in a steamer refining factories stuffed in side paper like material reconstituted from<br />

recycled <strong>of</strong> tobacco fines. It contains in addition to nicotine several additives<br />

such as ammonium, polystyrene foam, glycerol, cocoa, licorice, microorganism<br />

(bacteria and fungi from soil), pesticides, carbon dioxide and sugar.<br />

Nicotine in tobacco stimulates chemical reaction in the brain that leads to light<br />

feeling <strong>of</strong> pleasure being similar to natural body substances such as endorphin<br />

and dopamine’s, contrary to cocaine and heroin which induce deep temporary<br />

feeling <strong>of</strong> pleasure.(5)<br />

Epidemiology <strong>of</strong> tobacco<br />

More than one billion people smoke tobacco globally, smoking related diseases<br />

killed one in 10 adult, every 8 seconds someone dies from tobacco use.<br />

15 billion cigarettes are sold daily that make 10 million cigarettes are sold every<br />

minute. Smoking habits is very common all over the world; in Cambodia, China,<br />

Korea 67% <strong>of</strong> men smoke, in Philippines 60% <strong>of</strong> men smoke, in Japan 51% <strong>of</strong><br />

men smoke, half <strong>of</strong> all Malaysian are smokers.(6)<br />

Word Health Organization estimated tobacco smoking stratified by gender as<br />

depicted in Table 1.<br />

Table 1: Smoking Prevalence by Gender (2000, World Health Organization<br />

estimates) (6)<br />

Percent Smoking<br />

REGION MEN WOMEN<br />

Africa 29 4<br />

United States 35 22<br />

Eastern Mediterranean 35 4<br />

Europe 46 26<br />

Southeast Asia 44 4<br />

Western Pacific 60 8<br />

In the United States, smoking rates have dropped from 42% to 20.8% between<br />

1965 to 2006.(7)<br />

Unfortunately, there are no authenticating statistics about tobacco smoking in the<br />

<strong>Arab</strong> world apart from estimated statistics published by a different anti smoking<br />

charity organization. United State Census Bureau, International Data Base, 2004,<br />

present one <strong>of</strong> the most valuable data that gave Extrapolated Statistics in absence<br />

<strong>of</strong> solid data in the <strong>Arab</strong> World about tobacco smoking Table 2.(8)<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 21


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

Table 2: Extrapolated Statistics <strong>of</strong> smoking in some <strong>Arab</strong> countries (8)<br />

Country Estimated Smoker<br />

Egypt 16,974,184<br />

Gaza strip - West Bank 295,472-515,398<br />

Iraq 5,658,555<br />

Jordan 1,251,298<br />

Kuwait 503,433<br />

Lebanon 842,319<br />

Libya 1,255,843<br />

Saudi <strong>Arab</strong>ia 5,752,493<br />

Sudan 8,730,039<br />

Syria 4,017,762<br />

United <strong>Arab</strong> Emirates 562,833<br />

Yemen 4,465,545<br />

The following data was collected from 5 different sources could give impression<br />

about the magnitude <strong>of</strong> smoking in the Islamic world (Table 3)<br />

Country Population<br />

Million<br />

Table 3 : Smoking facts and figures in some Muslim countries<br />

Adult >15% Youth<br />

Prevalance <strong>of</strong><br />

tobacco use<br />

Adult<br />

Prevalance<br />

<strong>of</strong> tobacco<br />

smoking<br />

males %<br />

Adult<br />

Prevalance<br />

<strong>of</strong> tobacco<br />

smoking<br />

females %<br />

Adult<br />

Prevalance<br />

<strong>of</strong> tobacco<br />

smoking %<br />

Cigarette<br />

consumption<br />

(million sticks)<br />

Estimation<br />

Albania 3.6 77 NA 39.6 3.9 40.5 NA 2003<br />

Algeria 34 75 13.8 29.9 NA 29.9 21500 2003<br />

Bangladesh 141 64.5 5.8 48.6 25.4 36.8 23000 2004<br />

Bahrain 0.727 74.1 19.9 26.2 2.7 26.1 796 2001<br />

Egypt 83 69.7 12.6 59.3 2.7 29.9 61000 2005<br />

Indonesia 240 72 13.5 63.2 4.5 34.5 185000 2006<br />

Iran 66 79 13 24.1 4.3 14.2 48000 2005<br />

Iraq 29 62 20.3 25.7 1.9 25.8 NA 2006<br />

Jordan 6.3 69 17.2 61.7 7.9 62.7 NA 2002<br />

Kuwait 2.6 74 20.9 30 1.5 15.6 3216 2004<br />

Lebanon 4.01 74 28 29 6.9 29.1 6390 2002<br />

Libya 6.3 67 11.1 55.5 2.5 NA 5560 2003<br />

Malaysia 26 69 NA 43 1.6 26.4 22000 2003<br />

Morocco 35 70 14.5 29.5 0.3 29.5 14700 2006<br />

Oman 3.4 57 15.2 24.8 1 24.7 1776 2000<br />

Pakistan 176 63 10.1 32.4 5.7 19.1 59000 2003<br />

Palastine 3.9 56 39.9 NA NA 35.4 NA 2005<br />

Qatar 0.83 79 17.9 37 0.5 18.8 864 2005<br />

Saudi <strong>Arab</strong>ia 22 62 15.9 25.6 3.2 25.6 14431 2006<br />

Sudan 41 60 14 23.5 1.5 12.9 1475 2003<br />

Syria 20 64 35.5 50.6 9.9 44 10270 1999<br />

Tunisia 10 78 18.3 61.9 7.7 34.8 11498 2004<br />

Turkey 79 73 8.4 52 17.3 34.6 125000 2003<br />

UAE 4.7 80 24.9 27.2 2.4 26.1 3627 2003<br />

Yemen 24 54 21 77 29 44.5 5040 2006<br />

22 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info<br />

Year <strong>of</strong><br />

survey


Sources :<br />

CIA the 2008 world fact book 40<br />

WHO - Global Youth Tobacco Survey (GYTS) 41<br />

Country & Regional Pr<strong>of</strong>iles and Economics <strong>of</strong> Tobacco Briefs 42<br />

WHO Statistical Information System (WHOSIS) 43<br />

Nation Master 44<br />

In Saudi <strong>Arab</strong>ia there are no national data about smoking habits apart from dozens<br />

<strong>of</strong> studies describing smoking practice in universities and schools or smoking habits<br />

in health workers which might not describe the smoking problem in Saudi <strong>Arab</strong>ia,<br />

One <strong>of</strong> the studies based on questionnaires distributed among primary care clinic<br />

attendants that showed <strong>of</strong> 634 subjects, 34.4% (218) were current smokers, 16.4%<br />

(104) were ex-smokers, and 49.2% (312) were nonsmokers (10)<br />

Saudi anti smoking charity organization reported that six million smoker smoke<br />

15,000,000 000 cigarette annually costing more than one 1,000,000,000 Saudi<br />

Riyal based on import /export customs report, 23,000 person die annually as a<br />

direct result <strong>of</strong> cigarette smoking which means 63 person die on daily basis. Saudi<br />

Cancer Registry estimates that one third <strong>of</strong> cancer patient in Saudi <strong>Arab</strong>ia has<br />

smoking as causative agent.(11)<br />

Smoking Risk<br />

(Have you not reason then to be ashamed, and to forbeare this filthie noveltie, so<br />

basely grounded, so foolishly received and so grossly mistaken in the right use<br />

there<strong>of</strong>? In your abuse there<strong>of</strong> sinning against God, harming your selves both in<br />

persons and goods, and raking also thereby the marks and notes <strong>of</strong> vanity upon you:<br />

by the custom there<strong>of</strong> making your selves to be wondered at by all foreign civil<br />

nations, and by all strangers that come among you, to be scorned and contemned.<br />

A custom loathsome to the eye, hateful to the nose, harmful to the brain, dangerous<br />

to the lungs, and in the black stinking fume there<strong>of</strong>, nearest resembling the horrible<br />

Stigian smoke <strong>of</strong> the pit that is bottomless).<br />

The above quotation had been written more than 400 years ago by King James I<br />

<strong>of</strong> England in counter blasé to tobacco treaties in 1604. (12, 13)<br />

Tobacco smoking is one <strong>of</strong> the most common preventable health hazards that kill<br />

millions <strong>of</strong> peoples every year more than death caused by AIDS, suicide, murder,<br />

fires, accidental poisoning and traffic accidents combined.<br />

There are hundreds <strong>of</strong> studies correlate tobacco smokings to certain diseases<br />

which circulating around three major health problems: cardiovascular diseases,<br />

respiratory system diseases and cancer.<br />

The small fine particles that sneak through alveolar wall and exert their effects<br />

on the heart and blood vessels. Carbon Monoxide (CO) in tobacco impairs the<br />

hemoglobin ability to carry oxygen and the heart compensates for the low oxygen<br />

content by pumping more blood which is evident by increasing the heart rate by<br />

at least 30 % during smoking.(12)<br />

Smoking increases the platelet production, blood pressure (13), cholesterol and<br />

fibrinogen which together increase the risk <strong>of</strong> thrombi formation, arteriosclerosis<br />

and ischemia which could be fatal.<br />

Chronic obstructive pulmonary disease (COPD), defined as the ratio <strong>of</strong> forced<br />

expiratory volume in 1 second (FEV1) to the forced vital capacity (FVC) being <<br />

0.7 and the FEV1 being 50-80% <strong>of</strong> the expected value, caused mainly by smoking<br />

and it is a common cause <strong>of</strong> death in smokers.(14)<br />

Burning <strong>of</strong> organic compounds that occur during smoking releases several<br />

carcinogenic agents that damage the DNA causing several mutations. These<br />

agents contain hydrocarbons and acrolein which are potent carcinogens and<br />

as a result <strong>of</strong> that, smoking is linked to 15 different cancers and causes 30% <strong>of</strong><br />

cancer related death.<br />

Cancer <strong>of</strong> lips, oral cavity, larynx, pharynx, lung, pleura, esophagus, breast,<br />

stomach, kidney, pancreas, colon, cervix, acute myeloid leukemia and cancer <strong>of</strong><br />

bladder are well linked to tobacco smoking.<br />

Tobacco smoking contributes to other diseases such as cataracts, gum disease,<br />

aortic aneurism, and sudden infant death. It worsens diabetes complications and<br />

delay the wound healing.(15)<br />

Second Hand Smoking<br />

Many <strong>of</strong> 3800 compounds in tobacco smoke are known carcinogens. These<br />

compounds unfortunately do not just affect the smoker only but everyone around<br />

him who inhale this smoke.<br />

The passive smoker exposed to the side stream smoke emitted from the burning<br />

tip <strong>of</strong> the cigarette. Side stream smoke is hazardous because it contains high<br />

concentrations <strong>of</strong> ammonia, benzene, nicotine, carbon monoxide, and many<br />

carcinogens. Nonsmokers chronically exposed to side stream are believed to<br />

assume health risks similar to those <strong>of</strong> a light smoker. Children <strong>of</strong> parents who<br />

smoke have more respiratory infections, more hospitalizations for bronchitis and<br />

pneumonia, and a smaller rate <strong>of</strong> increase in lung function compared to children<br />

<strong>of</strong> parents who do not smoke, particularly during the first year <strong>of</strong> life. (16)<br />

Secondhand smoking or passive smoking or what is called “environmental tobacco<br />

exposure (ETS)” can cause the same problems as direct smoking. It is well known<br />

that non-smokers exposed to cigarette smoke in the workplace have an increased<br />

lung cancer risk that shown by the meta-analyses published by a Sasco AJ et al<br />

where lifelong non smokers with partners who smoke at home have a 20 to 30%<br />

greater risk <strong>of</strong> lung cancer than non-smokers who live with non-smokers.(17)<br />

Boyl P et al published data about non-smokers exposed to cigarette smoke in<br />

the workplace that showed increased lung cancer risk <strong>of</strong> 16 to 19% among non<br />

smokers. (18)<br />

Having a spouse who currently smokes was associated with an increased risk<br />

<strong>of</strong> first stroke among never-smokers (hazard ratio=1.42, 95% CI=1.05, 1.93)<br />

and former smokers (hazard ratio=1.72, 95% CI=1.33, 2.22). Former smokers<br />

married to current smokers had a stroke risk similar to respondents who themselves<br />

smoked.(19)<br />

A recent paper from China estimated the burden <strong>of</strong> diseases in adults from passive<br />

tobacco smoking for two major diseases: lung cancer and ischemic heart disease<br />

(IHD). It showed that passive smoking caused more than 22,000 lung cancer<br />

deaths in 2002 and when the toll <strong>of</strong> disability is added to that <strong>of</strong> mortality, passive<br />

smoking was responsible for the loss <strong>of</strong> nearly 230,000 years <strong>of</strong> healthy life from<br />

lung cancer. They estimated approximately 33,800 IHD deaths could be attributable<br />

to passive smoking in China in 2002. Passive smoking is also responsible for the<br />

loss <strong>of</strong> more than one quarter <strong>of</strong> a million years <strong>of</strong> healthy life from IHD.(20)<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 23


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

These data encouraged the health authority in many countries to implement<br />

smoking ban policy<br />

Smoking Cessation Benefits<br />

Smoking cessation is associated with important benefits such as improved lung<br />

function and a better general health and performance status. It might lead to longer<br />

survival and reduced complications <strong>of</strong> radiation therapy in lung cancer patient.(21)<br />

Costa F. et al tried to answer the question whether smoking cessation is worthwhile<br />

in patients with lung cancer as a common theme among patients that it “may be<br />

it is not worthwhile” given the bad prognosis. However, Costa found that: “it is<br />

worthwhile”.(22)<br />

(Smoking can influence lung cancer in several ways such as promoting relapse<br />

and the development <strong>of</strong> other types <strong>of</strong> cancer, smoking increases the growth and<br />

aggressiveness <strong>of</strong> tumors, nicotine interacts with the non-neuronal nicotinicacetylcholine<br />

receptors, leading to an increase in VGEF, nitric oxide and<br />

prostacyclin, inducing an increase in tumor growth, complicating surgery because<br />

smoking increases the surgical risk and postoperative complications, slowers wound<br />

healing and reduces survival after surgery, interfering with radiotherapy hence the<br />

hypoxia induced by carbon monoxide may be responsible for a worse response to<br />

radiotherapy. Smoking also increases the risk <strong>of</strong> radiation pneumonitis by promoting<br />

inflammation and diminishing the mucociliary clearance. As for chemotherapy,<br />

smoking interferes with cytochrome P450, accelerating the metabolism <strong>of</strong> several<br />

drugs including some chemotherapeutic agents (taxans, vinorelbine, etoposide,<br />

doxorubicin, gefitinib), diminishing their serum levels and their efficacy. It seems<br />

by this group <strong>of</strong> evidence that smoking cessation in lung cancer patients is in fact<br />

worthwhile.(22)<br />

Smoking cessation was associated with a decrease in the risks <strong>of</strong> ischemic stroke,<br />

subarachnoid hemorrhage, and MI.(23)<br />

Quitting smoking is associated with a substantial reduction in risk <strong>of</strong> all-cause<br />

mortality among patients with CHD, 36% reduction in crude relative risk (RR)<br />

<strong>of</strong> mortality for patients with CHD who quit compared with those who continued<br />

smoking.This risk reduction appears to be consistent regardless <strong>of</strong> age, sex, index<br />

cardiac event, country, and year <strong>of</strong> study commencement.(24)<br />

Smoking reduction was associated with a significant decrease in the risk <strong>of</strong> lung<br />

cancer. (25)<br />

Song YM, et al reported recently that smoking cessation substantially reduces the<br />

risk <strong>of</strong> smoking-related cancers <strong>of</strong> the lung, larynx, esophagus, and pancreas, and<br />

is the most effective method for reducing the risk <strong>of</strong> cancer among smokers.(25),<br />

Oral health usually improved after two weeks <strong>of</strong> tobacco cessation with resolution<br />

<strong>of</strong> smoking related oral lesion with overall improvement <strong>of</strong> periodontal and oral<br />

health.(26)<br />

Smoking Bans and Public Policies<br />

Smoking ban aims at protecting people from the harmful effects <strong>of</strong> second-hand<br />

smoke. Smoking ban policy is important tools in lowering smoking rates and<br />

promoting public health in addition lowering health service cost and improves<br />

morale and productivity and lower the overall cost <strong>of</strong> labor in a community.<br />

Effective tobacco control policies include law issuing, bans/restrictions on smoking<br />

in public areas and workplaces, increasing taxes on tobacco products, bans on<br />

advertising <strong>of</strong> tobacco products, land warning labels on cigarette boxes.<br />

The World Health Organization had published several policies and laws that helps<br />

health authority to implement cigarette smoking ban that helps many country to<br />

control the smoking habit and smoking becomes more difficult.<br />

Admissions for heart attacks dropped by 27% in Colorado after 18 months <strong>of</strong><br />

implementation. Similar results were seen in Ireland, Scotland, UK and Sweden.<br />

These results support the smoking ban fans.<br />

The European Union (EU) has been active in tobacco control policy since 1985<br />

when the Milan Council announced its intention to establish a Europe <strong>Against</strong><br />

Cancer (EAC) Programme. Shortly after the establishment <strong>of</strong> the EAC, first<br />

action plan the European Commission presented its first legislative proposals on<br />

tobacco control. Three <strong>of</strong> these proposals on labeling and maximum tar yields<br />

became Directives by 1992. The fourth on tobacco advertising finally became law<br />

in 1998 and is currently being transposed into national law in the 15 EU Member<br />

States. In 1996 the Commission published a Communication on the future <strong>of</strong> EU<br />

tobacco control and in 1999 at the 2nd European Conference on Tobacco and<br />

Health, the Social Affairs Commissioner announced his intention to bring forward<br />

further legislative proposals to amend an consolidate existing EU legislation in<br />

this sector. This article is intended to present an overview <strong>of</strong> EU tobacco control<br />

legislation from 1970 until 1998 and to look at future options post year 2000.(27)<br />

The smoking ban policies announced in Saudi <strong>Arab</strong>ia on the year 1984 with<br />

Royal order banning smoking in all governmental buildings. Unfortunately<br />

the implementation <strong>of</strong> this policy was very weak and people continue smoking<br />

breaking the royal order. More than 43 charity organizations started to be active<br />

in smoking ban campaign that result <strong>of</strong> increasing the taxation <strong>of</strong> cigarettes twice<br />

in the last tow decade and community based clinics started to receive smokers<br />

seeking help for smoking cessation maneuvers.<br />

Impact <strong>of</strong> religious rulings (Fatwa) on smoking:<br />

The Qur'ān, The holy book <strong>of</strong> Islam, does not specifically prohibit or denounce<br />

smoking directly, but gives behavioral guidance:<br />

“Don't throw yourself into danger by your own hands...” (Surat Al-Bakara 2/195)<br />

“You may eat, drink, but not waste” (Surat Al-A‘râf 7/31)<br />

Several Fatwa issued in most <strong>of</strong> Islamic countries announcing that tobacco smoking<br />

is unlawful activity “Haram” (sin) based on understanding <strong>of</strong> The Holy Quran<br />

that stated in Surat Al Araf : (The Prophet) who will enjoin upon them the doing<br />

<strong>of</strong> what is right, forbid them the doing <strong>of</strong> what is wrong, make lawful to them<br />

the good things <strong>of</strong> life, prohibit for them the evil things, and lift from them their<br />

burdens and the shackles that were (previously) upon them.(28)<br />

Smoking spoils a person's acts <strong>of</strong> worship and reduces their rewards, it spoils the<br />

prayer, which is the pillar <strong>of</strong> Islam. Allah's Messenger said: Whoever eats garlic<br />

or onion let him avoid us and our masjid, and stay in his home. The angels are<br />

surely hurt by things that hurt the human beings.(29)<br />

But although claimed that Islamic rules helped in smoking cessation program<br />

especially in Ramadan where the smoking is unlawful during the day time during<br />

fasting but few medical studies addressed this issue.<br />

24 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


One study conducted in University <strong>of</strong> Southern California, Los Angeles, California,<br />

USA. examines religious and cultural influences with adolescents' susceptibility to<br />

smoking among Muslim <strong>Arab</strong>-American adolescents and found religious influence<br />

and perceived negative consequences <strong>of</strong> smoking were protective against ever<br />

smoking for both genders (OR=0.7, 95% CI=0.5-0.9; OR=0.8, 95% CI=0.7-0.9,<br />

respectively). (30)<br />

Egyptian Smoking Prevention Research Institute conducted a survey in a rural<br />

village and two nearby schools in Qalyubia Governorate to assess the knowledge<br />

about religious ruling (Fatwa) and its impact on the quit attempts. The results<br />

showed that the majority <strong>of</strong> respondents (81 %) knew about the Fatwa on smoking.<br />

97.3% <strong>of</strong> all participants thought that smoking is a sin. This indicates a successful<br />

outreach program targeting mainly men through mosques.<br />

Radwan et al concluded that knowledge about the Fatwa on smoking or belief that<br />

smoking is a sin had no significant effect on quit attempts (31) but that definitely<br />

depends on personal believes and religious practice and obedience that varies<br />

from person to person and from society to other,<br />

In Pakistan, a study tried to see the effect <strong>of</strong> Islamic practice status <strong>of</strong> person and<br />

correlate that with their smoking habits to see the rule <strong>of</strong> Islam on prevention <strong>of</strong><br />

smoking and they found out <strong>of</strong> the study group 325 (32.5%) were found to be<br />

indulged in different kinds <strong>of</strong> smoking and concluded that Islam has a positive<br />

effect on smoking prevention.(32)<br />

Lung cancer prevention programs<br />

Lung cancer prevention programs depend on smoking avoidance. It is well known<br />

that cigarette smoking causes lung cancer and therefore smoking avoidance should<br />

result in decreased mortality from primary lung cancer.<br />

Nicotine replacement therapies in a form <strong>of</strong> gum, batch, spray, lozenge help to<br />

some extent in smoking-cessation programs and same apply to antidepressant<br />

therapy. The choice <strong>of</strong> therapy should be individualized based on the patient<br />

experience, preference and the agent side effects.<br />

The available data indicate that the intervention didn’t work for heavy-smokers<br />

where the quit rates was not significantly improved but it had improved among<br />

light-to-moderate smokers.<br />

Chemoprevention trial did not show promising result and is not yet established<br />

in standard clinical practice.<br />

Lung cancer screening for heavy-smokers is not yet established as standard clinical<br />

practice because no major benefits <strong>of</strong> cancer screening in lowering the morbidity<br />

or mortality <strong>of</strong> lung cancer in most <strong>of</strong> published screening program data.<br />

Smoking cessation programs<br />

Nancy Rigotti had published a practical guidelines for smoking cessation depending<br />

on pharmacotherapy and counseling which are the two approaches showing good<br />

result. Each is effective by itself, but the two in combination achieve the highest<br />

rates <strong>of</strong> smoking cessation.(33)<br />

Counseling<br />

Counseling about smoking cessation can be delivered effectively in person or<br />

by telephone. Group or individual counseling is effective when it is provided<br />

by trained counselors and includes repeated contacts over a period <strong>of</strong> at least<br />

four weeks. The efficacy <strong>of</strong> this approach increases as the amount <strong>of</strong> time spent<br />

with the patient increases. Cognitive behavioral methods form the core <strong>of</strong> most<br />

counseling programs. Typically, smokers learn to identify smoking cues, then use<br />

cognitive and behavioral methods to break the link between the cues and smoking.<br />

They also learn strategies for coping with stress, managing symptoms <strong>of</strong> nicotine<br />

withdrawal, and once they quit, preventing relapse, such as anticipating tempting<br />

situations and rehearsing coping strategies. (33-35)<br />

Smoking-counseling strategies are also summarized in pamphlets and booklets,<br />

audiotapes, videotapes, and computer programs. Written self-help material has<br />

negligible efficacy when used alone but may augment other interventions. Self-help<br />

material is more effective when its content is tailored to an individual patient's<br />

specific concerns or readiness to change(33-35)<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 25


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

Figure 1: The 5As Model for Smoking Cessation<br />

Ask<br />

All patients including<br />

adolescents about smoking<br />

status.<br />

Do you smoke?<br />

Advise<br />

to quit smoking<br />

to quit Assess smoking<br />

the interest to quit<br />

smoking Do you<br />

feel you want to quit?<br />

Y ES 
<br />

Assist<br />

Help the patient to quit smoking<br />

(STARR )<br />

(Setting a quit date,<br />

Telling friend,<br />

Anticipate challenges,<br />

Remove tobacco from patient<br />

environment,<br />

Recommend pharmacotherapy<br />

Arrange<br />

follow-up visit immediately after<br />

quit date<br />

Figure 1: The 5As Model for Smoking Cessation<br />

NEXT VISIT<br />

No/Not Now<br />

No/Not now<br />

Relapsed<br />

26 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info<br />

No



Figure 2: Sites <strong>of</strong> action <strong>of</strong> pharmacotherapy for smoking cessation.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 27


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

Table 4: Mechanism <strong>of</strong> action, common adverse effects, and efficacy in meta-analyses <strong>of</strong> pharmacotherapy’s <strong>of</strong> current or<br />

Pharmacotherapy Mechanism<br />

<strong>of</strong> action<br />

Nicotine patch Nicotine<br />

replacement<br />

therapy<br />

Nicotine gum Nicotine<br />

replacement<br />

therapy<br />

Nicotine lozenge Nicotine<br />

replacement<br />

therapy<br />

Nicotine inhaler Nicotine<br />

replacement<br />

therapy<br />

Nicotine nasal<br />

spray<br />

Nicotine<br />

replacement<br />

therapy<br />

future use for treating tobacco smoking dependence.<br />

Daily Dose Common adverse<br />

Effects<br />

14-25 mg , One patch daily<br />

for 4 weeks , decreased by<br />

7 mg every 2 weeks for 8<br />

weeks<br />

4-mg gum for 25 or more<br />

cigarettes per day and<br />

2-mg gum for less than 25<br />

cigarettes per day.<br />

Use should not exceed 24<br />

pieces per day<br />

4 mg for those who have<br />

their first cigarette within<br />

30 minutes <strong>of</strong> waking<br />

2 mg for those who have<br />

their first cigarette later<br />

than 30 minutes after<br />

waking.<br />

1 lozenge every 1 to 2<br />

hours for 6 weeks ; no<br />

fewer than 9 per day but no<br />

more than 20 per day. Use<br />

up to 12 weeks.<br />

1-2 doses per hour<br />

increasing as needed.<br />

Minimum recommended<br />

dosage is 8 doses per<br />

day; maximum is 40.<br />

(approximately 100 doses<br />

per bottle) 3 to 6 months<br />

with Gradual reduction<br />

One spray in each nostril<br />

(0.5 mg) 1-2 doses /hr ,<br />

Max 40/day for 3-6 months<br />

with gradual reduction.<br />

skin irritation insomnia,<br />

vivid dreams<br />

Nausea, vomiting,<br />

headache, dizziness,<br />

cold sweat, pallor,<br />

and weakness are<br />

all symptoms <strong>of</strong> an<br />

overdose<br />

Burning taste<br />

Hiccups<br />

Gastrointestinal<br />

symptoms, nausea<br />

Temporomandibular<br />

tenderness<br />

Tachycardia<br />

Mouth pain<br />

Hiccups<br />

Coughing<br />

Heartburn<br />

Sore throat<br />

Headache<br />

Indigestion<br />

Nausea<br />

Insomnia<br />

Irregular heartbeat<br />

Nausea, vomiting,<br />

headache, dizziness,<br />

cold sweat, pallor,<br />

and weakness are<br />

all symptoms <strong>of</strong> an<br />

overdose.<br />

Nasal airway reactions<br />

Dyspepsia<br />

Nausea, vomiting,<br />

headache, dizziness,<br />

cold sweat, pallor,<br />

and weakness are<br />

all symptoms <strong>of</strong> an<br />

overdose.<br />

Dependency - 15-<br />

20% use longer than<br />

recommended; 5% use<br />

at a higher dose than<br />

recommended<br />

Nasal irritation,<br />

Sneezing ,cough, teary<br />

eyes<br />

Efficacy<br />

Almost twice as<br />

effective as placebo<br />

when used 6-14 weeks<br />

at a dose <strong>of</strong> 14-25 mg<br />

(Fiore et al., 2008).39<br />

Long-term abstinence<br />

rates are increased<br />

approximately 50% over<br />

placebo (Fiore et al.,<br />

2008). .39<br />

The continuous<br />

abstinence rates at<br />

6 months compared<br />

to placebo are<br />

approximately double<br />

(24.2 vs. 14.4 for 2 mg<br />

dose and 23.6 vs. 10.2<br />

for 4 mg (Fiore et al,<br />

2008)39<br />

Long-term<br />

abstinence rates more<br />

approximately doubled<br />

when compared to<br />

placebo.39<br />

double the likelihood <strong>of</strong><br />

long-term quitting than<br />

those on placebo (Fiore<br />

et al., 2008).39<br />

28 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


Incentives for Smoking Cessation<br />

In recent randomized clinical trial, K Volpp et al found that financial incentives for<br />

smoking cessation significantly increased the rates <strong>of</strong> smoking cessation among<br />

2 matching smoker working in the same company in US where they randomly<br />

assigned 878 employees <strong>of</strong> a multinational company based in the United States<br />

to receive information about smoking-cessation programs (442 employees) or to<br />

receive information about programs plus financial incentives (436 employees).<br />

The financial incentives were $100 for completion <strong>of</strong> a smoking-cessation<br />

program, $250 for cessation <strong>of</strong> smoking within 6 months after study enrollment,<br />

as confirmed by a biochemical test, and $400 for abstinence for an additional 6<br />

months after the initial cessation, as confirmed by a biochemical test.<br />

The incentive group had significantly higher rates <strong>of</strong> smoking cessation than did<br />

the information-only group 9 or 12 months after enrollment (14.7% vs. 5.0%,<br />

P


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

9. N Ghouri, M Atcha, A Sheikh -BMJ 2006;332:291-294 (4 February),<br />

doi:10.1136/bmj.332.7536.291<br />

10. Siddiqui S, Ogbeide DO, Al Khalifa I. Smoking in a Saudi community:<br />

prevalance, influencing factors, and risk perception. Fam Med. 2001<br />

May;33(5):367-70.<br />

11. Saudi Antismoking Society Annual Report 2007.<br />

12. Haldane J (1895). "The action <strong>of</strong> carbonic oxide on man". J Physiol 18:<br />

430–462<br />

13. Krzyszt<strong>of</strong> Narkiewicz; Sverre E. Kjeldsen; Thomas Hedner , "Is smoking a<br />

causative factor <strong>of</strong> hypertension? "Blood pressure 14, (2 April 2005 ), 69 - 71<br />

14. Graham Devereux, ABC <strong>of</strong> chronic obstructive pulmonary disease Definition,<br />

epidemiology, and risk factors, BMJ 2006;332:1142-1144<br />

15. The Surgeon General (Health Consequences <strong>of</strong> Smoking, Surgeon General’s<br />

Report) US government 2004.<br />

16. Eriksen MP, LeMaistre CA, Newell GR. Health hazards <strong>of</strong> passive smoking,<br />

Annu Rev Public Health. 1988;9:47-70<br />

17. Sasco AJ, Secretan MB, Straif K. (2004). "Tobacco smoking and cancer: a<br />

brief review <strong>of</strong> recent epidemiological evidence". Lung Cancer 45 (Suppl 2):<br />

S3–9.<br />

18. Boyle P, Autier P, Bartelink H et al.. "European Code <strong>Against</strong> Cancer and<br />

scientific justification: third version (2003)". Ann Oncol. 14 (7).<br />

19. Glymour MM, Defries TB, Kawachi I, Avendano M. Spousal smoking and<br />

incidence <strong>of</strong> first stroke: the Health and Retirement Study. Am J Prev Med. 2008<br />

Sep;35(3):245-8<br />

20. Gan Q, Smith KR, Hammond SK, Hu TW ,Disease burden <strong>of</strong> adult lung<br />

cancer and ischaemic heart disease from passive tobacco smoking in China. Tob<br />

Control. 2007 Dec;16(6):417-22<br />

21. Nieder C, Bremnes RM,Effects <strong>of</strong> Smoking Cessation on Hypoxia and its<br />

Potential Impact on Radiation Treatment Effects in Lung Cancer Patients.<br />

Strahlenther Onkol. 2008 Nov;184(11):605-609. Epub 2008 Nov 19<br />

22. Costa F, Marques A, Figueiredo F, Barata F,Smoking cessation in lung cancer<br />

patients: Is it worthwhile? Rev Port Pneumol. 2006 Dec;12(6 Suppl 1):49-50<br />

23. Song YM, Cho HJ.Risk <strong>of</strong> stroke and myocardial infarction after reduction<br />

or cessation <strong>of</strong> cigarette smoking: a cohort study in korean men. Stroke. 2008<br />

Sep; 39(9):2432-8. Epub 2008 Jul 10<br />

24. Critchley JA, Capewell S. Mortality risk reduction associated with smoking<br />

cessation in patients with coronary heart disease: a systematic review. JAMA.<br />

2003 Jul 2;290(1):86-97<br />

25. Song YM, Sung J, Cho HJ, Reduction and cessation <strong>of</strong> cigarette smoking<br />

and risk <strong>of</strong> cancer: a cohort study <strong>of</strong> Korean men. J Clin Oncol. 2008 Nov 1;<br />

26(31):5101-6. Epub 2008 Oct 6<br />

26. Shibly O, Cummings KM, Zambon JJ. Resolution <strong>of</strong> oral lesions after<br />

tobacco cessation. J Periodontol. 2008 Sep;79(9):1797-801<br />

27. Godfrey F , An overview <strong>of</strong> European Union tobacco control legislation.<br />

Cent Eur J Public Health. 2000 May;8(2):128-31<br />

28. Al-A'raf 7:157. Holy Quran<br />

29. Al-Bukhaari and Muslim from Jabir and other Sahaabah<br />

30. Islam SM, Johnson CA. Ethn Health. 2003 Nov;8(4):319-37.<br />

31. Radwan et al , J Egypt Soc Parasitol. 2003 Dec;33(3 Suppl):1087-101.<br />

32. Hameed A, Jalil MA, Noreen R, Mughal I, Rauf S. J Ayub Med Coll<br />

Abbottabad. 2002 Jan-Mar;14(1):23-5.<br />

33. Nancy A. Rigotti, Treatment <strong>of</strong> Tobacco Use and Dependence, NEJM Feb<br />

14,2003,7:346:506-512<br />

34. Fiore MC, Bailey WC, Cohen SJ, et al. Treating tobacco use and dependence.<br />

Rockville, Md.: Department <strong>of</strong> Health and Human Services, Public Health<br />

Service, 2000. (Also available at http://www.surgeongeneral.gov/tobacco.)<br />

35. Lancaster T, Stead L, Silagy C, Sowden A. Effectiveness <strong>of</strong> interventions<br />

to help people stop smoking: findings from the Cochrane Library. BMJ<br />

2000;321:355-358<br />

36. Laura Carrozzi, Francesco Pistelli and Giovanni Viegi Review:<br />

Pharmacotherapy for smoking cessation Therapeutic Advances in Respiratory<br />

Disease 2008; 2; 301)<br />

37. K.G. Volpp and Others, A Randomized, Controlled Trial <strong>of</strong> Financial<br />

Incentives for Smoking Cessation,NEJM,360:699-709 ,Feb 12, 2009 No7<br />

38. Fiore MC, Bailey WC, Cohen SJ, et al. Treating tobacco use and dependence.<br />

Rockville, Md.: Department <strong>of</strong> Health and Human Services, Public Health<br />

Service, 2000<br />

39. Fiore, M.C., Jae´n, C.R., Baker, T.B., Bailey, W.C.,Benowitz, N.L., Curry,<br />

S.J. et al. (2008)Treating Tobacco Use and Dependence: 2008 Update.Clinical<br />

Practice Guideline. Rockville, MDUS Department <strong>of</strong> Health and Human<br />

Services,Public Health Service. May 2008.<br />

40. https://www.cia.gov/library/publications/the-world-factbook/<br />

41. http://www.who.int/infobase/report.<br />

42. http://www.worldbank.org/tobacco/countrybrief.asp<br />

43. http://www.who.int/whosis/en/<br />

44. http://www.nationmaster.com<br />

30 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


THE USE OF MRI FOR THE EARLY DETECTION OF BREAST CANCER<br />

Comstock C. 1<br />

(1) University <strong>of</strong> California, San Diego, Ca, USA<br />

Corresponding Author: Dr. Christopher Comstock, MD<br />

Associate Clinical Pr<strong>of</strong>essor <strong>of</strong> Radiology, Director <strong>of</strong> Breast Imaging<br />

University <strong>of</strong> California, San Diego, La Jolla, CA, United States <strong>of</strong> America<br />

Dynamic contrast enhanced MRI <strong>of</strong> the breast is not limited by breast density<br />

and has been shown to be extremely sensitive in the detection <strong>of</strong> invasive breast<br />

cancer. However, because <strong>of</strong> the variable sensitivity <strong>of</strong> MRI for ductal carcinoma<br />

in situ (DCIS), between 45% and 100%, MRI is currently not recommended as a<br />

replacement for screening mammography(1). A more recent single institution study<br />

suggests that MRI may have a higher sensitivity as compared to mammography<br />

for DCIS than previously thought, particularly for high-grade DCIS(2). The use<br />

<strong>of</strong> MRI in the general population has been limited by its moderate specificity,<br />

leading to false positives and unnecessary biopsies. Therefore, its use has been<br />

focused on studying patients in whom the yield from MRI is likely to be higher.<br />

Multiple studies have shown that MRI is a useful tool as an adjuvant to screening<br />

mammography in women at high risk for breast cancer(3-5).<br />

In April 2007 the American Cancer Society (ACS) published new guidelines for<br />

screening high-risk woman(6). Risk is greatest for women with genetic mutations<br />

including BRCA1, BRCA2, p53, Cowden’s, and those women who underwent<br />

radiation therapy for lymphoma at an early age. Methods for estimating risk<br />

based on medical and family history include the Gail, Claus, and BRCAPRO<br />

mathematical models. Women who have a 20-25% lifetime risk for breast cancer<br />

based on family history as calculated by any <strong>of</strong> the aforementioned models are<br />

also considered high risk and appropriate for annual MRI. Women whose benefit<br />

from screening MRI is considered questionable by the ACS due to insufficient data<br />

include; women with a personal history <strong>of</strong> breast cancer, prior biopsy yielding atypia<br />

or women with extremely dense breasts on mammography(6,7). The decision to<br />

perform screening MRI in these women should be made on a case by case basis.<br />

Table: Risk factors that place women at increased risk <strong>of</strong> breast cancer<br />

BRCA1 or BRCA2 mutation, or untested first degree relative <strong>of</strong> known<br />

carrier<br />

Chest radiation between age 10-30 for Hodgkin’s Lymphoma<br />

Lifetime risk <strong>of</strong> 20-25% as determine by statistical risk assessment models<br />

such as BRCAPRO or Gail based on personal and family history<br />

Other genetic mutations including p53 and Cowden<br />

References<br />

1. Bazzocchi M, Zuiani C, <strong>Pan</strong>izza P, et al. Contrast-enhanced breast MRI in<br />

patients with suspicious microcalcifi cations on mammography: results <strong>of</strong> a<br />

multicenter trial. AJR Am J Roentgenol 2006; 186: 1723–32.<br />

2. Kuhl CK, Schrading S, Bieling HB, et al. MRI for diagnosis <strong>of</strong> pure ductal<br />

carcinoma in situ: a prospective observational study. Lancet. 2007 Aug<br />

11;370(9586):485-92.<br />

3. Kriege M, Brekelmans CT, Boetes C, et al. Efficacy <strong>of</strong> MRI and<br />

mammography for breast-cancer screening in women with a familial or genetic<br />

predisposition. N Engl J Med. 2004 Jul 29;351(5):427-37.<br />

4. Lehman CD, Blume JD, Weatherall P, et al. Screening women at high risk for<br />

breast cancer with mammography and magnetic resonance imaging. Cancer<br />

2005;103:1898–905.<br />

5. Kuhl CK, Schrading S, Leutner CC, et al. Mammography, breast ultrasound,<br />

and magnetic resonance imaging for surveillance <strong>of</strong> women at high familial<br />

risk for breast cancer. J Clin Oncol. 2005 Nov 20;23(33):8469-76.<br />

6. Port ER, Park A, Borgen PI, Morris E, Montgomery LL. Results <strong>of</strong> MRI<br />

screening for breast cancer in high-risk patients with LCIS and atypical<br />

hyperplasia. Ann Surg Oncol. 2007 Mar;14(3):1051-7. Epub 2007 Jan 7.<br />

7. Saslow D, Boetes C, Burke W, et al. American Cancer Society guidelines for<br />

breast screening with MRI as an adjunct to mammography. CA Cancer J Clin.<br />

2007 Mar-Apr;57(2):75-89.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 31


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

DIET, PHYSICAL ACTIVITY, AND OBESITY IN THE PREVENTION<br />

AND RECURRENCE OF BREAST CANCER: RELEVANCE TO SAUDI<br />

ARABIAN WOMEN<br />

Mc Tiernan, A.<br />

Corresponding Author: Anne McTiernan, MD, PhD<br />

Director, Prevention Center, Fred Hutchinson Cancer Research Center<br />

PO Box 19024, , M4-B874, Seattle, Washington, USA 98109<br />

E-mail: amctiern@fhcrc.org<br />

Abstract<br />

Objectives: Diet, physical activity, and overweight/obesity can alter risk <strong>of</strong><br />

developing breast cancer and may affect prognosis among women who are<br />

diagnosed with breast cancer. This manuscript outlines lifestyle behavioral<br />

strategies that show promise in the prevention and treatment/rehabilitation <strong>of</strong><br />

breast cancer.<br />

Methods: Literature was summarized regarding the following major lifestyle<br />

behaviors that are relevant to breast cancer etiology and control: physical activity,<br />

weight control, and diet.<br />

Results: Compared with sedentary women, the risk <strong>of</strong> developing breast cancer is<br />

30 – 40% lower for women who exercise at moderate to vigorous levels for 3 – 4<br />

hours per week. Women who are overweight or obese have a 30 – 50% increased<br />

risk for postmenopausal breast cancer development compared with normalweight<br />

women. In contrast, overweight and obesity decrease risk <strong>of</strong> breast cancer<br />

occurring during the premenopausal years. There is no evidence that any one dietary<br />

component is related to breast cancer risk, although recent evidence suggests that<br />

women with low vitamin D levels may be at increased risk. Breast cancer patients<br />

at any age have increased risk <strong>of</strong> poor survival if they are overweight, obese, or<br />

sedentary. One large randomized trial found that a low-fat dietary pattern improved<br />

disease-free survival in women with early stage breast cancer.<br />

Conclusions: To reduce breast cancer risk, and to improve prognosis in women<br />

with breast cancer, weight should be maintained at normal levels (body mass index<br />

< 25.0 kg/m2) by reducing calorie intake and increasing physical activity (at least<br />

150 minutes/week). The preferred dietary pattern for maintaining normal weight<br />

is low in fat, high in vegetables and fruits, and low in refined carbohydrates. A<br />

low-fat dietary pattern may improve prognosis in breast cancer patients.<br />

Introduction<br />

Diet, physical activity, and overweight/obesity can alter risk <strong>of</strong> developing breast<br />

cancer and may affect prognosis among women who are diagnosed with breast<br />

cancer. Globally, rates <strong>of</strong> breast cancer incidence vary widely by geographic area.<br />

Only a small part <strong>of</strong> these differences are due to genetic differences, few chemical<br />

or other carcinogen exposures have been linked to risk, and the remainder <strong>of</strong> cases<br />

are likely due to individual health and lifestyle behaviors.(1) Around the world,<br />

within-country changes over time in breast cancer incidence have been paralleled<br />

by significant lifestyle and health behavior changes.(2) Experimental animal<br />

studies provide confirmation <strong>of</strong> an observable effects <strong>of</strong> overweight/obesity, diet,<br />

and physical activity on breast biology.(1)<br />

The International Agency for Research on Cancer (IARC) estimates that 25%<br />

<strong>of</strong> breast cancer cases worldwide are due to overweight/obesity and a sedentary<br />

lifestyle.(1) Recently, an expert panel commissioned by the American Institute for<br />

Cancer Research and the World Cancer Research Fund estimated that recommended<br />

diets, together with maintenance <strong>of</strong> physical activity and appropriate BMI, can in<br />

time reduce breast cancer incidence.(3) (4)<br />

An American Cancer Society cohort study <strong>of</strong> 495,477 women followed for 16 years<br />

found that risk <strong>of</strong> breast cancer mortality increased significantly with increasing<br />

level <strong>of</strong> obesity; compared with women with a body mass index (BMI) under 25.0<br />

kg/m2, those with a BMI <strong>of</strong> 25 – 29.9 kg/m2, 30 – 34.9 kg/m2, 35 – 39.9 kg/m2,<br />

and >40 kg/m2 had a relative risk <strong>of</strong> breast cancer mortality <strong>of</strong> 1.34, 1.63, 1.7, and<br />

2.12, respectively.(5) Therefore, lifestyle changes to increase physical activity and<br />

reduce excess weight might be expected to have a major impact on breast cancer<br />

risk and mortality. In countries where routine mammogram or other screening is<br />

not generally available, it is even more important to promote lifestyle changes to<br />

reduce risk <strong>of</strong> breast cancer and to improve prognosis in women with breast cancer.<br />

This manuscript outlines lifestyle behavioral strategies that show promise in the<br />

prevention and treatment/rehabilitation <strong>of</strong> breast cancer. It covers the following<br />

major lifestyle behaviors that are relevant to breast cancer etiology and control:<br />

physical activity, weight control, and diet. While alcohol use has been consistently<br />

linked to increased risk for breast cancer (6) likely through its effects on estrogen<br />

metabolism, it is very unlikely to be consumed by Saudi women, and therefore<br />

alcohol use is not included in this manuscript.<br />

Lifestyle Factors and Breast Cancer Prevention<br />

Physical Activity<br />

There is a very large body <strong>of</strong> epidemiologic data on the association between<br />

exercise and breast cancer.(7) Over twenty published cohort studies (8-30) have<br />

investigated the association between physical activity and risk <strong>of</strong> breast cancer,<br />

<strong>of</strong> which most (8-31) showed evidence <strong>of</strong> a reduced risk for breast cancer in<br />

women who were classified at the highest vs. lowest levels <strong>of</strong> physical activity.<br />

The reduction in risk ranged from 10 to 70 percent for the most active women,<br />

and on average was 30 – 40% lower for women who exercised at moderate to<br />

vigorous levels for 3 – 4 hours per week. The definition <strong>of</strong> “most active” varied<br />

by study, and depended on the questions asked, the population studied, and the<br />

researchers’ choice <strong>of</strong> categories <strong>of</strong> amount <strong>of</strong> activity.<br />

In the United States’ Women’s Health Initiative Observational Study, we studied<br />

the association between physical activity and risk <strong>of</strong> postmenopausal breast cancer.<br />

(18) Exercise reduced the risk <strong>of</strong> all types <strong>of</strong> breast cancer regardless <strong>of</strong> receptor<br />

hormone status or stage <strong>of</strong> disease, although the numbers in some subgroups were<br />

too small to precisely determine risk estimates. The Women’s Health Initiative<br />

Observational Study is a cohort <strong>of</strong> more than 93,000 women from across the United<br />

States, including women from varying race and ethnic populations. Moderate<br />

activities such as walking were as protective against breast cancer as were vigorous<br />

activities such as jogging. The beneficial effect <strong>of</strong> exercise was most pronounced<br />

in women with body mass index less than 27 kg/m2.<br />

In a study <strong>of</strong> over 25,000 Norwegian women, decreasing risk for breast cancer<br />

with increasing levels <strong>of</strong> leisure time physical activity (p trend = .08), and<br />

physical activity at work (p trend = .004) was observed.(27) There have been two<br />

publications from the U.S. Nurses’ Health Study cohorts on the association between<br />

physical activity and risk <strong>of</strong> breast cancer: one study assessed recreational activity<br />

reported at just one point in time,(24) and the other assessed repeated measures<br />

<strong>of</strong> activity at several time points during follow-up.(23) While the former study<br />

observed no association between physical activity and risk for breast cancer, the<br />

latter found that women who engaged in an average <strong>of</strong> seven or more hours per<br />

week <strong>of</strong> physical activity had an 18 percent lower chance (95% confidence interval<br />

32 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


3% to 30%) <strong>of</strong> developing breast cancer compared with women who engaged in<br />

less than one hour per week <strong>of</strong> such activities. Activity other than leisure-time<br />

activity was not assessed, which may obscure physical activity effects, as there<br />

may be considerable effect <strong>of</strong> occupational activity in a population <strong>of</strong> nurses.<br />

Some cohort studies have estimated breast cancer risk according to participation in<br />

college sports, some studied occupational physical activity only, several examined<br />

recreational exercise only, and others assessed effects <strong>of</strong> both occupational and<br />

recreational physical activity. Methods <strong>of</strong> measuring physical activity differed<br />

between studies, ranging from simply asking subjects questions such as, “In your<br />

usual day, aside from recreation, how active are you?”, to a physician-administered<br />

questionnaire that ascertained how many hours per day a subject usually spent<br />

sleeping, resting, sedentary, or at slight, moderate, or heavy activities, to detailed<br />

question <strong>of</strong> current and history <strong>of</strong> regular participation in various sports and other<br />

recreational exercise activities at different life periods. Therefore, it is even more<br />

significant that such a high proportion <strong>of</strong> studies have found an association between<br />

increased physical activity and reduced risk for breast cancer.<br />

More than two dozen case-control studies have been published on the association<br />

between physical activity and risk <strong>of</strong> breast cancer,(7) <strong>of</strong> which more than three<br />

quarters support a reduced risk for breast cancer in women who were the most<br />

active compared with sedentary women. Reduction in risk ranged from 10% to 70%.<br />

Reduced risks associated with increased physical activity have been observed<br />

for both premenopausal and postmenopausal breast cancer. Data from around<br />

the world suggest that physical activity is associated with reduced risk for breast<br />

cancer in women <strong>of</strong> diverse races and ethnicities.(30, 32-36)<br />

It is not clear at what ages physical activity provides the most protection against<br />

breast cancer. In a small number <strong>of</strong> case-control studies,(37-39) lifetime leisure<br />

activity was ascertained, while in other studies, activity levels at adolescence and<br />

discrete adult periods were obtained. Some studies found a reduction in risk with<br />

increased physical activity in adolescence, while in other studies risk reduction<br />

was limited to adult physical activity.<br />

It is important in studying the association between physical activity and breast<br />

cancer to control for potentially confounding factors, to be sure that the association<br />

between physical activity and breast cancer is not due to extraneous factors.<br />

Most studies have controlled for potential confounding factors such as age,<br />

reproductive history, and body mass index, and some have also adjusted for<br />

patterns <strong>of</strong> dietary intake. Since body size and adiposity may lie in the causal<br />

pathway between physical activity and breast cancer risk, simple adjustment may<br />

not give a complete picture. Indeed, some investigators found that breast cancer<br />

risk reduction associated with level <strong>of</strong> physical activity was limited to the leanest<br />

women.(27, 39-41) Adjustment for dietary macronutrient intake or caloric intake<br />

did not confound the physical activity-breast cancer relationship in studies that<br />

assessed dietary patterns.(15, 27, 33, 41-44)<br />

Identifications <strong>of</strong> the mechanisms linking physical activity and cancer risk will help<br />

in determining optimum ages to exercise, and the dose, frequency, and intensity<br />

<strong>of</strong> physical activities needed to protect against breast cancer.<br />

Early menarche (before age 12), increased numbers <strong>of</strong> ovulatory cycles, late first<br />

birth or nulliparity, lack <strong>of</strong> lactation, late age at menopause, increased number <strong>of</strong><br />

lifetime ovulatory cycles, increased interval between menarche and menopause,<br />

and high concentrations <strong>of</strong> endogenous sex hormones (estrogens, testosterone)<br />

have been found to increase risk <strong>of</strong> breast cancer from 20 percent to more 400<br />

percent.(45, 46) Several <strong>of</strong> these reproductive and hormonal factors are affected<br />

to some degree by physical activity.<br />

In observational studies, pre-teen and teenage girls participating in vigorous<br />

activities such as ballet dancing, gymnastics, and running have been noted to<br />

experience a high incidence <strong>of</strong> primary and secondary amenorrhea, delayed<br />

menarche, and more irregular cycles, compared with non-athlete girls.(47, 48) A<br />

cross-sectional study <strong>of</strong> 174 girls aged 14 – 17 years found that girls who expended<br />

600 or more kcal energy per week (comparable to two or more hours per week<br />

in activities such as aerobic exercise classes, swimming, jogging, or tennis) were<br />

two to three times more likely than less active girls to have anovulatory menstrual<br />

cycles.(49)<br />

Exercise during reproductive life has been shown to alter the concentrations <strong>of</strong><br />

sex hormones in human intervention studies.(50, 51) A high intensity exercise<br />

intervention in 28 untrained college women with normal ovulation resulted in<br />

reversible abnormal luteal function in two-thirds and loss <strong>of</strong> luteinizing hormone<br />

surge in more than 50% <strong>of</strong> subjects.(51) The greatest menstrual changes were<br />

observed during the periods <strong>of</strong> most intense training, and in those women who<br />

had been randomized to a weight loss (vs. weight maintenance) group. It appears,<br />

therefore, that a low body weight in addition to increased exercise is required to<br />

reduce ovulation. Thus, intense prolonged exercise or caloric restriction, or some<br />

combination <strong>of</strong> these, may be required.<br />

Postmenopausal women produce estrogen through the peripheral conversion (mainly<br />

in fat cells) <strong>of</strong> adrenal androgens to estrogens.(52) In postmenopausal women,<br />

increased physical activity is associated with decreased serum concentrations<br />

<strong>of</strong> estradiol, estrone, and androgen,(53-55) and increased concentrations <strong>of</strong> sex<br />

hormone binding globulin.(56) One published randomized clinical trial has shown<br />

that a moderate exercise program reduces endogenous sex hormones in overweight/<br />

obese sedentary postmenopausal women.(57, 58) Cross-sectional data from the<br />

Women’s Health Initiative show that both a sedentary lifestyle and increased<br />

body weight are significantly related to increased blood levels <strong>of</strong> estrogens in<br />

postmenopausal women.(59)<br />

Weight and Body Composition<br />

Postmenopausal Breast Cancer<br />

Body mass index (BMI) is the most common measure <strong>of</strong> adiposity used in studies<br />

<strong>of</strong> weight, adiposity, and breast cancer. Obesity experts have developed the<br />

following categories <strong>of</strong> adiposity based on BMI: underweight (30.0 kg/m2). (60)<br />

More than 100 studies have reported on the association <strong>of</strong> weight or BMI at<br />

different ages, central fat distribution, or adult weight gain and risk <strong>of</strong> breast<br />

cancer incidence.(61) Taken together, these studies found that women who are<br />

overweight or obese have a 30 – 50% increased risk for postmenopausal breast<br />

cancer development compared with normal-weight women. In contrast, overweight<br />

and obesity decrease risk <strong>of</strong> breast cancer occurring during the premenopausal<br />

years. The Women’s Health Initiative Observational Study is a multi-ethnic, multisite<br />

cohort study <strong>of</strong> women aged 50 – 79 at study entry.(62) Women underwent<br />

several clinic measures <strong>of</strong> weight and body mass when entering the cohort,<br />

including height, weight, waist, and hip circumferences, and self-reported lifetime<br />

weight history. Among who had never used menopausal hormone therapy, women<br />

with a BMI >= 31.1 kg/m2 had a statistically significant 2.5 times greater risk <strong>of</strong><br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 33


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

developing breast cancer compared with women whose BMI was 22.6 kg/m2 or<br />

less (63). The Nurses’ Health Study also observed a 60 percent increased risk for<br />

postmenopausal breast cancer associated with overweight and obesity in women<br />

who had never use menopausal hormone therapy.(64) Results <strong>of</strong> case-control<br />

studies are similar to those <strong>of</strong> cohort studies.(61)<br />

Premenopausal Breast Cancer<br />

The association <strong>of</strong> weight and adiposity with risk for premenopausal breast cancer<br />

differs from that in postmenopausal breast cancer. In particular, weight or BMI<br />

are slightly inversely related to premenopausal breast cancer risk. In a recent<br />

meta-analysis <strong>of</strong> BMI and cancer incidence (65) that included 20 epidemiologic<br />

studies <strong>of</strong> BMI and premenopausal breast cancer, the overall summary relative risk<br />

<strong>of</strong> premenopausal breast cancer for each 5 kg/m2 increase in BMI was 0.92 (95%<br />

CI 0.88-0.97, p < 0.001). A recent study <strong>of</strong> 113 130 premenopausal participants<br />

(with 1398 cases <strong>of</strong> incident breast cancer) in the Nurses’ Health Study II cohort<br />

reported a significant linear inverse trend between current BMI and breast cancer<br />

incidence (P < 0.001) that was not explained by menstrual cycle characteristics or<br />

infertility due to an ovulatory disorder (covariate- adjusted hazard ratio for breast<br />

cancer in women with a BMI > 30 kg/m2 vs 20.0-22.4 kg/m2, 0.81; 95% CI, 0.68-<br />

0.96). (66) BMI at age 18 years was the strongest predictor <strong>of</strong> breast cancer risk<br />

(covariate-adjusted hazard ratio for breast cancer in women with a BMI at age<br />

18 years > 27.5 kg/m2 vs 20.0-22.4 kg/m2 was 0.57; 95% CI, 0.41-0.81). The<br />

inverse association <strong>of</strong> BMI with premenopausal breast cancer risk was limited to<br />

hormone receptor positive breast cancers.<br />

These data on BMI and premenopausal breast cancer is particularly relevant to <strong>Arab</strong><br />

women in the Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia, where the median age at breast cancer<br />

diagnosis is 50 years, likely representing a high proportion <strong>of</strong> cases developing<br />

in the premenopausal years (67). It is not clear why premenopausal breast cancer<br />

differs from postmenopausal breast cancer with respect to adiposity, but one<br />

hypothesis is that obese girls, teenagers, and premenopausal women have fewer<br />

ovulatory cycles and therefore are exposed to lower levels <strong>of</strong> ovarian hormones<br />

including estrogens, progesterone, and testosterone compared with normal-weight<br />

girls and young women (68).<br />

Adult Weight Gain<br />

Weight gain during adulthood has been consistently associated with increased<br />

risk for postmenopausal breast cancer.(63, 64, 69-76) Findings from two large<br />

cohort studies suggest that the doubling <strong>of</strong> risk associated with a gain in BMI<br />

from age 18 greater than 9.7 kg/m2 (Women’s Health Initiative) or a weight gain<br />

great than 20 kg (Nurses’ Health Study) is limited to women who had never used<br />

postmenopausal menopausal hormone therapy.(63, 64) In these studies, a 20%<br />

increase in risk was observed for BMI gains between 3.5 – 6.2 kg/m2 (Women’s<br />

Health Initiative) or weight gains between 2 to 20 kg (Nurses Health Study).<br />

Increased adult weight gain has also been found to be a consistent predictor <strong>of</strong><br />

increased risk for breast cancer in case-control studies.(61)<br />

Abdominal Fat<br />

High levels <strong>of</strong> abdominal fat have been associated with up to a doubling <strong>of</strong> breast<br />

cancer risk among postmenopausal women in a small number <strong>of</strong> cohort studies<br />

compared low levels <strong>of</strong> abdominal fat,(63, 69, 72, 77, 78) independent <strong>of</strong> BMI. In<br />

the Women’s Health Initiative, statistically significant trends <strong>of</strong> increasing breast<br />

cancer risk with increasing waist and hip circumferences were observed among<br />

women who had never used menopausal hormone therapy.(63) Women in the<br />

highest quintile <strong>of</strong> either waist or hip circumference had approximately twice the<br />

risk for breast cancer development compared with women in the lowest quintile. In<br />

the Nurses’ Health Study, the relative risk among women in the highest vs. lowest<br />

quintile <strong>of</strong> waist circumference was 1.2 among women overall and 1.9 among women<br />

who had never used menopausal hormone therapy.(64) However, not all studies<br />

have found an association <strong>of</strong> abdominal circumference with breast cancer risk.<br />

Effect <strong>of</strong> Intentional Weight Loss<br />

There are few data on the association <strong>of</strong> weight loss and breast cancer risk. In<br />

three studies, weight loss occurring over a prolonged interval was associated with<br />

a non-statistically significant reduced risk.(69, 70, 79) In another study, weight<br />

loss in the decade before diagnosis was also associated with a non-statistically<br />

significant decreased risk.(76) One study in premenopausal women aged 44<br />

years or less found a statistically significant 36 percent decreased risk with<br />

weight loss from age 20 to interview that was limited to cases with low-grade<br />

tumors.(80) One study in postmenopausal women aged 50 – 74 years found a<br />

statistically significant 24 percent decreased risk <strong>of</strong> breast cancer with weight loss<br />

from age 18 to interview.(74) In an analysis in the Nurses' Health Study, 87,143<br />

postmenopausal women, aged 30 to 55 years and free <strong>of</strong> cancer were followed<br />

for up to 26 years (1976 – 2002) to assess effects <strong>of</strong> lifetime weight change on<br />

breast cancer risk. (81) A total <strong>of</strong> 4393 cases <strong>of</strong> invasive breast cancer were<br />

documented during this follow-up. Compared with those who maintained weight,<br />

women who gained 25.0 kg or more from age 18 years had a 45 percent increased<br />

risk <strong>of</strong> breast cancer that was statistically significant. Among women who had<br />

never taken menopausal hormone therapy, those who gained 25 kg. or more had<br />

almost a doubling <strong>of</strong> risk <strong>of</strong> breast cancer compared with those who remained<br />

weight stable. Compared with weight maintenance, women who gained 10.0 kg<br />

or more after menopause had a statistically significant 18 percent increased risk<br />

<strong>of</strong> breast cancer occurrence. Women who had never used menopausal hormone<br />

therapy, lost 10.0 kg or more since menopause, and kept the weight <strong>of</strong>f had less<br />

than half the risk <strong>of</strong> breast cancer compared with those who maintained weight,<br />

and the result was statistically significant. The researchers estimated that 15.0%<br />

<strong>of</strong> breast cancer cases in the Nurses’ Health Study cohort could be attributable to<br />

weight gain <strong>of</strong> 2.0 kg or more since age 18 years and 4.4% could be attributable<br />

to weight gain <strong>of</strong> 2.0 kg or more since menopause. Among those who did not use<br />

menopausal hormone therapy, these attributable risks were 24.2% for a weight<br />

gain since age 18 years and 7.6% for weight gain since menopause.<br />

The majority <strong>of</strong> the studies on weight and breast cancer risk have been conducted<br />

in European and North American populations. Nevertheless, the available data<br />

suggest that increased adiposity increases risk for breast cancer across geographic,<br />

cultural, race, and ethnic groups.(2)<br />

Mechanisms<br />

There are several likely mechanisms linking increased adiposity to risk for<br />

postmenopausal breast cancer. After menopause, adipose tissue is the main site<br />

<strong>of</strong> estrogen production through aromatization <strong>of</strong> androgens to estrogens.(52)<br />

Overweight and obese postmenopausal women have high concentrations <strong>of</strong> estrone,<br />

estradiol, testosterone and low concentrations <strong>of</strong> sex hormone binding globulin,<br />

compared with leaner women.(55) Testosterone concentrations are increased in<br />

both premenopausal and postmenopausal overweight/obese women compared<br />

with leaner women, perhaps due to increased conversion <strong>of</strong> androstenedione to<br />

testosterone in adipose tissue.(82) In a recent study using a random sub sample <strong>of</strong><br />

women in the Women’s Health Initiative Dietary Modification Trial, women with<br />

a high BMI and low self-reported physical activity, had higher levels <strong>of</strong> estrone,<br />

estradiol and free estradiol, and lower levels <strong>of</strong> sex-hormone binding globulin<br />

(SHBG) than women a similar BMI who were active as well as those with low<br />

BMI in either activity category.(59)<br />

34 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


Insulin promotes cancer cell growth, and therefore could explain part <strong>of</strong> the link<br />

between increased adiposity and breast cancer risk, because overweight and<br />

obese women have increased blood insulin levels compared with normal-weight<br />

women.(83) Insulin-like growth factors (IGFs) stimulate cell turnover in most<br />

body tissues, and have been associated with increased risk <strong>of</strong> breast cancer.(84)<br />

IGF is down-regulated by increased production <strong>of</strong> its binding protein (IGFBP-1),<br />

which can result from increased exercise, decreased caloric intake, and decreased<br />

body weight.(85, 86) Decreased IGF activity may increase the hepatic synthesis<br />

<strong>of</strong> sex hormone binding globulin, resulting in diminished availability <strong>of</strong> free sex<br />

hormone. Thus, increased exercise could result in lowered biologically available<br />

endogenous sex hormones via a cascade <strong>of</strong> metabolic and hormonal events, and<br />

thus a lowered risk <strong>of</strong> breast cancer.<br />

Diet<br />

The large international and temporal variations in risk for breast cancer prompted<br />

interest in possible dietary causes <strong>of</strong> breast cancer.(3) Several studies examined<br />

particular international and inter-cultural dietary differences, and proposed that diets<br />

that are low in fat and high in fruits, vegetables, fiber, and complex carbohydrates<br />

might lower risk for breast cancer.(87, 88) Although prospective observational<br />

studies in humans have generally not supported this hypothesis, several animal<br />

experimental studies have provided support for an association between certain<br />

dietary patterns and reduced risk for breast cancer.(89) Animal experimental studies<br />

have shown increased development (via promotion <strong>of</strong> tumorigenesis) <strong>of</strong> mammary<br />

tumors with increased polyunsaturated or saturated fats intake.(3) However, it is<br />

not clear if the dietary fat per se, or the increased energy intake, was responsible<br />

for the increased development <strong>of</strong> mammary tumors in these studies. Indeed, recent<br />

animal model evidence shows that both caloric restriction and increased exercise<br />

reduce mammary tumor development (90, 91)<br />

Human experiments have tested the effects <strong>of</strong> low fat and high fiber diets on some<br />

breast cancer biomarkers such as mammographic density and blood estrogens.<br />

The effect <strong>of</strong> a low-fat, high vegetable and fruit diet on mammographic density<br />

was tested in a randomized clinical trial in 817 women.(92) Women randomized<br />

to the diet arm experienced a 6.1% decrease in mammographic density over two<br />

years compared with a 2.1% decrease in controls (p = 0.01). Several clinical trials<br />

have shown a reduction in circulating estrogen levels with institution <strong>of</strong> a low-fat<br />

dietary pattern,(93) although there is still a question regarding whether it was the<br />

diet per se, or the weight loss resulting from the dietary changes, that caused the<br />

reduction in hormones.(94)<br />

The Women’s Health Initiative Dietary Modification clinical trial, begun in<br />

1993, included over 48,000 postmenopausal women aged 50 – 79 from diverse<br />

geographic, cultural, race and ethnic groups around the United States.(95) Women<br />

were randomly assigned to diet modification (40%) or comparison group (60%).<br />

The dietary modification goal was 20% calories from fat, 5 servings <strong>of</strong> fruits/<br />

vegetables per day, and 6 servings <strong>of</strong> grains per day. During follow-up, dietary fat<br />

intake was significantly lowered in the dietary modification group compared with<br />

the comparison group. The difference between groups in change from baseline<br />

for percentage <strong>of</strong> energy from fat varied from 10.7% at year 1 to 8.1% at year 6.<br />

Vegetable and fruit consumption was higher in the intervention group by at least<br />

1 serving per day. Over the 8.1-year average follow-up period, a total <strong>of</strong> 655<br />

women developed invasive breast cancer in the intervention group and 1072 in<br />

the comparison group, which resulted in a nine percent reduction in risk in women<br />

in the diet group versus control that was not statistically significant. Additional<br />

analyses suggested a lower risk among women who reduced dietary fat intake to<br />

the greatest degree, provided evidence <strong>of</strong> risk reduction among women having<br />

a high-fat diet at baseline, and suggested a dietary effect that varies by hormone<br />

receptor characteristics <strong>of</strong> the tumor.<br />

While increased intake <strong>of</strong> dietary fat per se has not been established as a risk<br />

factor for breast cancer, increased dietary fat typically increases caloric intake.<br />

This results in overweight and obesity, which are consistent risk factors for<br />

postmenopausal breast cancer. Therefore, prudent advice for women wanting to<br />

avoid lifetime weight gain, overweight, and obesity, would be to follow a diet<br />

that is low in dietary fat.<br />

Vegetables and Fruits<br />

Early epidemiologic studies suggested an association <strong>of</strong> increased intake <strong>of</strong><br />

vegetables and fruits with decreased risk for breast cancer.(3) A combined analysis<br />

<strong>of</strong> eight cohort studies representing 351,825 women (7377 breast cancer cases),<br />

however, found no association between intake <strong>of</strong> vegetable and fruits and risk<br />

<strong>of</strong> breast cancer.(96)<br />

Soy, Is<strong>of</strong>lavones, and Lignans<br />

Epidemiologic studies indicate that increased consumption <strong>of</strong> soy products is<br />

associated with reduced risk for breast cancer.(66, 93) Many soy-based foods are<br />

available, including t<strong>of</strong>u, soy milk, soy cheeses, frozen “yogurt,” breakfast shakes,<br />

soy nuts, meat substitutes, and salad dressings.(97) Recent evidence suggests,<br />

however, that genistein, one component <strong>of</strong> soy, may promote the growth <strong>of</strong> some<br />

estrogen-sensitive tumors and reduce the efficacy <strong>of</strong> tamoxifen. This emphasizes<br />

the need for additional studies to determine whether soy is safe for women with<br />

breast cancer or who are at high risk for breast cancer.(98, 99)<br />

Phytoestrogens can act as weak estrogens and as estrogen antagonists, depending<br />

on the hormonal status <strong>of</strong> the woman. Thus, increased phytoestrogen intake such<br />

as soy can compete with endogenous estrogens in premenopausal women, and<br />

reduce overall estrogen exposure to the breast. Conversely, phytoestrogens can<br />

increase estrogen activities in women with low endogenous levels <strong>of</strong> estrogens, e.g.<br />

postmenopausal women, and thereby increase the breast’s exposure to estrogen.<br />

These findings have been observed in animal experiments and in a small number<br />

<strong>of</strong> human experimental studies.(3, 100-102)<br />

Meat and Dairy<br />

Some studies have suggested that increased intake <strong>of</strong> meats increases risk for<br />

breast cancer development, but other studies have not supported this.(3) Part <strong>of</strong> the<br />

discrepancy in findings may be the different levels <strong>of</strong> carcinogens and mutagens<br />

included in meat in different areas around the world.<br />

Intake <strong>of</strong> dairy foods has not been found to be associated with risk for breast<br />

cancer, and a recent report from a large cohort study found that increased intake<br />

<strong>of</strong> low-fat dairy products in premenopausal women was associated with decreased<br />

risk <strong>of</strong> breast cancer.(103) In that study, the multivariable relative risks comparing<br />

more than 1 serving per day vs. 3 or fewer servings per month intake categories<br />

were 0.68 (95% CI = 0.55 to 0.86) for low-fat dairy foods and 0.72 (95% CI =<br />

0.56 to 0.91) for skim/low-fat milk. The fat content <strong>of</strong> high-fat dairy products may<br />

promote increased risk for breast cancer through increasing energy intake in the<br />

diet. On the other hand, the high calcium and vitamin D content <strong>of</strong> supplemented<br />

dairy products may be protective against breast cancer.(103)<br />

Vitamins and Minerals<br />

Several epidemiologic studies have investigated the association between dietary<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 35


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

and supplement intake <strong>of</strong> various vitamins and minerals, and risk <strong>of</strong> breast<br />

cancer. Specific micronutrients that have been associated with decreased risk<br />

include carotenoids, folate, calcium, vitamin D, lycopene, and vitamin C.(3,<br />

104-106) The studies have had mixed results, however, and because they have<br />

all been observational, are not conclusive. Recently, several studies have found<br />

an association between low blood levels <strong>of</strong> vitamin D and risk for breast cancer<br />

(107) Each <strong>of</strong> the studies categorized vitamin D levels differently, but overall it<br />

appears that women who had levels below the clinically “normal” levels had an<br />

increased relative risk for breast cancer compared with women with the highest<br />

blood levels. A protective effect <strong>of</strong> vitamin D, if substantiated, is relevant to Saudi<br />

<strong>Arab</strong>ia, where many <strong>of</strong> the women have low blood levels <strong>of</strong> vitamin D due to<br />

low sun exposure.<br />

Relevance to Women <strong>of</strong> the Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

Several <strong>of</strong> the dietary and lifestyle factors described above are relevant to women<br />

<strong>of</strong> the Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia. The prevalences <strong>of</strong> overweight/obesity in Saudi<br />

women is estimated to be over 75 percent (108). Physical activity levels in<br />

Saudi women are extremely low (109). Vitamin D levels in Saudi women are<br />

reportedly low (110), likely due to decreased sun exposure, obesity, and lack <strong>of</strong><br />

dietary supplementation. These lifestyle factors will likely result in an increased<br />

incidence <strong>of</strong> breast cancer in the future in Saudi <strong>Arab</strong>ia. Therefore, there are several<br />

nutritional and lifestyle factors that could be promoted among Saudi women to<br />

reduce risk for breast cancer:<br />

1. Reduce overweight and obesity and maintain a normal weight over the<br />

lifetime by reducing calorie intake and increasing physical activity. The<br />

preferred dietary pattern for maintaining normal weight is low in fat, high<br />

in vegetables and fruits, and low in refined carbohydrates.<br />

2. Engage in at least 150 minutes per week <strong>of</strong> aerobic physical activity. This<br />

could be done with brisk walking, with home stationary bikes, with active<br />

housework or gardening, with the use <strong>of</strong> home instructional videos, or<br />

in classes specially designed for women. Participation in athletics is not<br />

necessary for women to become physically active. Encouragement <strong>of</strong> gym and<br />

active play activities for girls would be beneficial for reducing breast cancer<br />

risk and for reducing incidence <strong>of</strong> overweight and obesity in young women.<br />

3. Increase vitamin D intake through supplementation. In the U.S. the<br />

recommended minimal daily intake <strong>of</strong> vitamin D3 is 400 – 1000 IU/day. In<br />

Saudi <strong>Arab</strong>ia, with low sun exposure in women, consideration for testing<br />

and supplementing vitamin D blood levels could have wide health benefits,<br />

as vitamin D is thought to reduce risk for several other chronic diseases.<br />

Lifestyle Factors and Prognosis in Women with Breast Cancer<br />

Once women develop breast cancer, they may be at increased risk <strong>of</strong> recurrence<br />

and poorer survival if they are overweight or obese. The effects <strong>of</strong> obesity on<br />

cancer outcome are substantial, where observed, and <strong>of</strong> potentially great clinical<br />

importance. The prevalence <strong>of</strong> overweight and obesity is higher in patients with<br />

some forms <strong>of</strong> cancer, compared with individuals from the general population<br />

(IARC). Compounding this is that weight gain after diagnosis is common in<br />

some breast cancer patients, especially among those receiving systemic adjuvant<br />

therapy. (111, 112) Weight gain during the post-breast cancer diagnosis period<br />

has also been associated with an adverse effect on recurrence risk and survival.<br />

(111) In addition to adversely affecting prognosis, overweight and obesity also<br />

increase the risk <strong>of</strong> several complications from cancer treatment, and increase<br />

risk <strong>of</strong> several co-morbidities. There are several potential mechanisms that might<br />

explain the link between increased adiposity and reduced prognosis, including<br />

hormonal, inflammatory, and immune system effects. Although definitive clinical<br />

trials testing weight loss effects on prognosis in breast cancer patients have not<br />

been conducted, strategies for weight control may be helpful for some breast<br />

cancer patients and survivors.<br />

Obesity and Breast Cancer Mortality – Non-Patient Populations<br />

In the American Cancer Society Prevention Study II, a prospective cohort study<br />

in 900,000 American adults, 57,145 cancer deaths were identified during 16 years<br />

<strong>of</strong> follow-up.(5) Cancer mortality was determined through personal inquiries and<br />

linkage with the National Death Index. The relative risks (RR) for breast cancer<br />

for increasing category <strong>of</strong> BMI, compared with women with BMI 40.0). The test for trend was highly significant (p <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


were measured in a fasting state prior to initiation <strong>of</strong> adjuvant treatment in 535<br />

women with T1-3, N0-1, M0 breast cancer. After a median 50 months follow-up,<br />

obesity predicted distant disease-free and overall survival (p


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

A recent study investigated whether activity undertaken prior to diagnosis<br />

influenced breast cancer survival in a population-based cohort <strong>of</strong> 1264 women<br />

ages 20 to 54 years who were diagnosed with invasive breast cancer between<br />

1990 and 1992 and followed for 8 to 10 years with 290 deaths. For women in the<br />

highest quartile <strong>of</strong> activity in the year before diagnosis compared with the lowest<br />

quartile, risk <strong>of</strong> dying from breast cancer was reduced by 22 percent. High activity<br />

was associated with a 30 percent reduced risk <strong>of</strong> death among women who were<br />

overweight or obese at the time <strong>of</strong> diagnosis that was statistically significant, but<br />

not among normal-weight or underweight women.(132)<br />

Dietary Composition and Breast Cancer Prognosis<br />

In the Women’s Intervention Nutrition Study (WINS), 2437 women with breast<br />

cancer were randomly assigned between 1994 – 2001 in a ratio <strong>of</strong> 40:60 to dietary<br />

intervention (n = 975) or control (n = 1462) groups and followed for a median<br />

60 months. Dietary fat intake was lower in the intervention than in the control<br />

group (33.3 versus 51.3 fat grams/day at 12 months, respectively), corresponding<br />

to a statistically significant (P = .005), 6-pound lower mean body weight in the<br />

intervention group. A total <strong>of</strong> 277 relapse events (local, regional, distant, or<br />

ipsilateral breast cancer recurrence or new contralateral breast cancer) were reported<br />

in 96 <strong>of</strong> 975 (9.8%) women in the dietary group and 181 <strong>of</strong> 1462 (12.4%) women<br />

in the control group. The hazard ratio <strong>of</strong> relapse events in the intervention group<br />

compared with the control group was 0.76 (95% CI = 0.60 to 0.98, P = .077 for<br />

stratified log rank and P = .034 for adjusted Cox model analysis). Exploratory<br />

analyses suggested a greater effect <strong>of</strong> the dietary intervention among women with<br />

hormone receptor negative tumors.(133)<br />

Potential Mechanisms for an Adverse Prognostic Effect <strong>of</strong> Obesity<br />

Several mechanisms have been proposed for an adverse prognostic effect <strong>of</strong><br />

obesity in cancer.(111) These mechanisms include increased levels <strong>of</strong> circulating<br />

hormones such as estrogen and androgens; reduced levels <strong>of</strong> sex hormone binding<br />

globulin (SHBG) which thereby increases the levels <strong>of</strong> free estradiol and free<br />

testosterone; increased levels <strong>of</strong> insulin and insulin-like growth factors; reduced<br />

levels <strong>of</strong> insulin-like growth factor binding globulin; increased levels <strong>of</strong> leptin;<br />

increased levels <strong>of</strong> cytokines; effects <strong>of</strong> diet; reduced immune functioning; and<br />

chemotherapy underdosing in obese patients. Although some biological evidence<br />

exists to support many <strong>of</strong> these potential mechanisms, there is little direct evidence<br />

<strong>of</strong> their role.<br />

Estrogens, Androgens, and Adiposity in Women<br />

Estrogens can promote growth <strong>of</strong> several hormone-dependent tumors, particularly<br />

breast and endometrium, and conversely, anti-estrogens or withdrawal <strong>of</strong><br />

endogenous estrogens are effective adjuvant treatments for breast cancer.<br />

(134, 135) Postmenopausal women produce estrogens in fat and other tissue<br />

through the aromatization <strong>of</strong> androgens to estrogens. The enzyme aromatase is<br />

abundantly present in adipose tissue, especially subcutaneous fat. Estrogens are<br />

tumor promoters in vitro and in vivo, and women with high circulating levels <strong>of</strong><br />

estrogens are at increased risk <strong>of</strong> developing breast cancer.(46)<br />

Postmenopausal women who are overweight or obese have elevated levels <strong>of</strong><br />

estrogens compared with lighter-weight women. In a population-based cohort <strong>of</strong><br />

505 postmenopausal women with Stage 0-3a breast cancer (the Health, Eating,<br />

Activity, Lifestyle – HEAL – Study), adiposity was positively and statistically<br />

significantly associated with circulating levels <strong>of</strong> estrone, estradiol, and free<br />

estradiol.(136) Women were recruited to this study through the Surveillance,<br />

Epidemiology, and End Results (SEER) cancer registries <strong>of</strong> Western Washington<br />

and New Mexico, and were primarily non-Hispanic and Hispanic Whites. Between<br />

4 – 12 months after diagnosis, anthropometric measures and blood draws were<br />

obtained on all women and DEXA scans were obtained on 415 women. Obese<br />

women (BMI >30) had 35% higher concentrations <strong>of</strong> estrone and 130% higher<br />

concentrations <strong>of</strong> estradiol, compared with lighter women (BMI <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


Abdominal Fat<br />

The body fat that is stored in the intra-abdominal area is thought to have special<br />

physiological properties, and is more associated than other body fat with risk<br />

factors for diabetes and cardiovascular disease. Increased intra-abdominal fat is<br />

associated with increased levels <strong>of</strong> insulin and total and LDL cholesterol.<br />

The role <strong>of</strong> visceral fat on cancer prognosis has not been established. Increased<br />

intra-abdominal fat, however, is associated with increased circulating levels <strong>of</strong><br />

serum insulin and glucose,(138) and as described above, insulin may be a tumor<br />

growth promoter. Therefore, interventions that decrease intra-abdominal fat levels<br />

may be hypothesized to improve cancer prognosis. In a small study, a 2-month<br />

low-fat diet and structured exercise intervention in women with a history <strong>of</strong> breast<br />

cancer resulted in a significant decrease in waist circumference.(139) In a clinical<br />

trial in 173 postmenopausal overweight-obese sedentary women without breast<br />

cancer, exercise significantly decreased intra-abdominal fat.(138)<br />

Obesity is also associated with inflammatory markers including C-reactive protein,<br />

serum amyloid A (SAA), interleukin-6 (IL-6), interleukin-1 (IL-1), and tumor<br />

necrosis factor-α (TNF-α), some <strong>of</strong> which been shown to be higher in patients<br />

with metastatic cancer compared with normals and with persons with early cancer.<br />

(111) Despite a paucity <strong>of</strong> data, it seems plausible that patients with depressed<br />

immune function might be at increased risk <strong>of</strong> tumor progression. Studies suggest<br />

decreased immune function in obese individuals, and increased immune function<br />

with exercise in breast cancer survivors.(140)<br />

Methods for Weight Loss and Maintenance for Breast Cancer Patients<br />

There are several methods for weight loss or control that have been tested in the<br />

general population(141) or in persons with other obesity-related conditions such as<br />

diabetes and cardiovascular disease.(142) Detailed guidelines for the identification,<br />

diagnosis, and treatment <strong>of</strong> overweight and obesity have been published by several<br />

institutes <strong>of</strong> the National Institutes <strong>of</strong> Health,(141) and recently from the U.S.<br />

Preventive Task Force.(143) There are no specific guidelines for weight loss or<br />

maintenance methods in breast cancer patients or survivors, however.<br />

The first step for clinicians should be the assessment <strong>of</strong> body composition including<br />

BMI.(143) For overweight (BMI 25.0 – 29.9 kg/m2) or obese (BMI >30 kg/m2)<br />

patients, physicians can then apply a weight loss strategy as outlined by the NHLBI<br />

Obesity Education Initiative Expert <strong>Pan</strong>el.(141) (The clinician should be aware<br />

that BMI does not always accurately reflect adiposity. Thus, for example, very<br />

muscular or athletic women may have a BMI above 25.0, without excess adipose<br />

tissue.) The NHLBI Obesity Education Initiative Expert <strong>Pan</strong>el recommends a<br />

weight loss treatment algorithm that combines dietary therapy, physical activity,<br />

and behavioral treatment, provided on an ongoing basis to promote weight loss<br />

and maintenance. For select patients, additional therapies such as medications<br />

and surgery could be considered.<br />

Behavioral Weight Loss Therapy<br />

Reviews <strong>of</strong> randomized trials in healthy obese individuals and in those at high risk<br />

for other diseases (e.g. pre-diabetics, hypertensives) show that the combination <strong>of</strong><br />

diet and behavioral treatment typically delivered in 15 to 24 weekly groups sessions,<br />

produces an average weight loss <strong>of</strong> approximately 8.5 kg (mean body weight<br />

reduction = 9%).(141) This degree <strong>of</strong> weight loss is associated with significant<br />

improvements in blood pressure, blood glucose and psychological well being. In<br />

the year following behavioral treatment participants regain typically 30% – 40%<br />

<strong>of</strong> their lost weight. However, relatively few studies have provided behavioral<br />

treatment lasting more than six months, and follow-up studies conducted 2 to 5<br />

years after behavioral treatment have documented a gradual return to baseline<br />

weights in most individuals. Long-term success is more likely when participants are<br />

provided with extended treatment programs. Support for the efficacy <strong>of</strong> extended<br />

lifestyle treatment has been well documented.(141)<br />

Weight Loss Diets<br />

The success <strong>of</strong> most dietary weight loss therapies has rested on reducing caloric<br />

intake below that required for current weight maintenance, e.g. creating a negative<br />

energy balance. A low-fat, reduced calorie diet has been shown to produce significant<br />

weight loss when combined with behavioral change counseling.(141) The first<br />

step in most weight loss diets is self-monitoring, where the individual records all<br />

food eaten each day. Then, the daily intake <strong>of</strong> calories, fat, fiber, carbohydrates,<br />

or all <strong>of</strong> these, can be tallied by the individual or weight loss counselor. Another<br />

key step is frequent and regular weighings by a health pr<strong>of</strong>essional or weight loss<br />

counselor. A major key to sustained weight loss is to achieve life-long dietary<br />

pattern changes, rather than short term “crash” diets.<br />

There is preliminary evidence that very-low-carbohydrate diets may work through<br />

additional mechanisms beyond simple calorie restriction, and produce greater<br />

weight loss compared with low calorie diets,(144, 145) although over a year, low<br />

carbohydrate diets may not be more efficacious than reduced calorie diets.(145)<br />

All diets have similar issues, in that most patients regain weight within a year<br />

after initial weight loss.(143)<br />

Weight Loss Pharmacotherapy<br />

For obese patients, or for those with BMI >27 with serious co-morbidities, weight<br />

loss medications may be a useful adjunct to diet and exercise therapy.(143, 146) Two<br />

medications are currently available in the United States for weight loss, and both<br />

are efficacious in the short term, although long-term efficacy data are not available.<br />

Sibutramine, a dopamine, norepinephrine, and serotonin reuptake inhibitor inhibits<br />

appetite through a central mechanism. In a review <strong>of</strong> 7 randomized clinical trials,<br />

sibutramine combined with lifestyle change promoted weight loss <strong>of</strong> 2.8 to 4.2<br />

kg over 8 to 52 weeks in healthy adults and those with controlled hypertension.<br />

(146) However, patients regained weight after cessation <strong>of</strong> treatment. Orlistat, a<br />

gastrointestinal lipase inhibitor, prevents fat absorption. In 10 randomized trials, it<br />

has produced an average 3.5 kg weight loss over 1 – 2 years, in excess <strong>of</strong> control<br />

(lifestyle alone) weight loss. A more recent review with updated trial data(143)<br />

concluded that therapy with sibutramine or orlistat combined with lifestyle change<br />

produced weight loss <strong>of</strong> 3 to 5 kg over that <strong>of</strong> control (lifestyle alone), and that<br />

prolonged use continued this weight loss over up to 2 years. Two other medication,<br />

phentermine and mazindol produced similar weight loss in the short term, but are<br />

not approved for long-term use. This same review concluded that three additional<br />

medications showed mixed results: metformin, diethylproprion, and fluoxetine.<br />

None <strong>of</strong> these medications have been specifically tested in breast cancer patients<br />

or survivors, and therefore their effects on prognosis or other aspects <strong>of</strong> the cancer<br />

experience are unknown.<br />

Bariatric Surgery<br />

For severely obese persons (BMI >40.0) or for patients with serious co-morbidities<br />

for whom obesity poses an extreme risk, more invasive methods <strong>of</strong> weight loss<br />

can be considered such as bariatric surgery.(147) The effect <strong>of</strong> weight loss surgery,<br />

however, has not been tested in persons who have had breast cancer, and is likely<br />

to only be appropriate for women with very early stage disease.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 39


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

Exercise and Weight Loss and Maintenance<br />

Physical activity may provide a low-risk method <strong>of</strong> preventing weight gain and<br />

promoting maintenance <strong>of</strong> weight loss in overweight and obese men and women.<br />

(148) Studies have shown losses in total body weight from exercise training<br />

without dieting.(123) In a review <strong>of</strong> several hundred studies <strong>of</strong> exercise training<br />

and weight loss, Wilmore concluded that the average weight loss over 12 months<br />

<strong>of</strong> exercise training would amount to only 3.2 kg.(149) Unlike weight-loss dietary<br />

interventions, physical activity also increases or maintains muscle mass and<br />

increases cardiorespiratory fitness levels.<br />

Weight Loss Studies in Breast Cancer Patients and Survivors<br />

Studies <strong>of</strong> weight loss reduction in breast cancer patients have been few and with<br />

mixed results. The Mayo Clinic randomized 107 breast cancer patients to monthly<br />

non-intensive dietician counseling. Median weight increase at six months was 2<br />

kg in the counseling group versus 3.5 kg in the control group, a non- significant<br />

difference.(150) Goodwin evaluated a multidisciplinary approach combining group<br />

dietary sessions, psychological support groups, and exercise programs in 61 breast<br />

cancer patients. For women with BMI >25 kg/m2, weight loss was 1.63 ± 4.22 kg,<br />

and aerobic exercise increase was a strong predictor <strong>of</strong> successful weight loss.(116)<br />

In a study <strong>of</strong> 34 obese breast cancer survivors, a combination <strong>of</strong> individualized<br />

counseling and Weight Watchers program produced greater weight loss than<br />

either alone or control.(151) Weight change after 12 months <strong>of</strong> intervention was:<br />

+0.85 kg in the control group, -2.6 kg in the Weight Watchers group, -8.0 kg in<br />

the individualized counseling group, and -9.4 kg in the comprehensive group that<br />

used both individualized counseling and Weight Watchers. Weight loss relative to<br />

control was statistically significant in the comprehensive group 3, 6, and 12 months<br />

after randomization, whereas weight loss in the individualized group was significant<br />

only at 12 months. The study resulted in weight loss <strong>of</strong> 10% or more <strong>of</strong> initial body<br />

weight in 6 <strong>of</strong> 10 women in the comprehensive group after 12 months. These same<br />

researchers found that the comprehensive group experienced significant declines<br />

in leptin and improvements in lipids.(152) de Waard and colleagues randomized<br />

102 postmenopausal women (median BMI 27 kg/m2) with a recent breast cancer<br />

diagnosis to a weight loss program involving step-wise reduction in caloric intake<br />

versus a control group. After one year, median weight loss was 6.0 kg with the<br />

intervention (p <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


2. McTiernan A. <strong>Association</strong>s between energy balance and body mass index and<br />

risk <strong>of</strong> breast carcinoma in women from diverse racial and ethnic backgrounds<br />

in the U.S. Cancer 2000;88(5 Suppl):1248-55.<br />

3. World Cancer Research Fund <strong>Pan</strong>el (Potter JD Chair). Food, Nutrition and the<br />

Prevention <strong>of</strong> Cancer: A Global Perspective. Washington, DC: American Institute<br />

for Cancer Research; 1997.<br />

4. Research WCRFAIfC. Food, Nutrition, Physical Activity, and the Prevention <strong>of</strong><br />

Cancer: A Global Perspective.; Washington DC: AICR, 2007.<br />

5. Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity,<br />

and mortality from cancer in a prospectively studied cohort <strong>of</strong> U.S. adults. N Engl<br />

J Med 2003;348(17):1625-38.<br />

6. Hamajima N, Hirose K, Tajima K, Rohan T, Calle EE, Heath CW, Jr., et al.<br />

Alcohol, tobacco and breast cancer—collaborative reanalysis <strong>of</strong> individual data<br />

from 53 epidemiological studies, including 58,515 women with breast cancer and<br />

95,067 women without the disease. Br J Cancer 2002;87(11):1234-45.<br />

7. Thune I, Furberg AS. Physical activity and cancer risk: dose-response and<br />

cancer, all sites and site-specific. Med Sci Sports Exerc 2001;33(6 Suppl):S530-50.<br />

8. Albanes D, Blair A, Taylor PR. Physical activity and risk <strong>of</strong> cancer in the<br />

NHANES I population. Am J Public Health 1989;79(6):744-50.<br />

9. Breslow RA, Ballard-Barbash R, Munoz K, Graubard BI. Long-term<br />

recreational physical activity and breast cancer in the National Health and<br />

Nutrition Examination Survey I epidemiologic follow-up study. Cancer Epidemiol<br />

Biomarkers Prev 2001;10(7):805-8.<br />

10. Calle EE, Murphy TK, Rodriguez C, Thun MJ, Heath CW, Jr. Occupation and<br />

breast cancer mortality in a prospective cohort <strong>of</strong> US women. Am J Epidemiol<br />

1998;148(2):191-7.<br />

11. Cerhan JR, Chiu BCH, Wallace RB, al. e. Physical activity, physical function,<br />

and the risk <strong>of</strong> breast cancer in a prospective study among elderly women. J<br />

Gerontol 1998;53A:M251-M256.<br />

12. Dallal CM, Sullivan-Halley J, Ross RK, Wang Y, Deapen D, Horn-Ross PL, et<br />

al. Long-term recreational physical activity and risk <strong>of</strong> invasive and in situ breast<br />

cancer: the California teachers study. Arch Intern Med 2007;167(4):408-15.<br />

13. Dirx MJ, Voorrips LE, Goldbohm RA, van den Brandt PA. Baseline recreational<br />

physical activity, history <strong>of</strong> sports participation, and postmenopausal breast<br />

carcinoma risk in the Netherlands Cohort Study. Cancer 2001;92(6):1638-49.<br />

14. Dorgan JF, Brown C, Barrett M, Splansky GL, Kreger BE, D'Agostino RB, et<br />

al. Physical activity and risk <strong>of</strong> breast cancer in the Framingham Heart Study.<br />

Am J Epidemiol 1994;139(7):662-9.<br />

15. Fraser GE, Shavlik D. Risk factors, lifetime risk, and age at onset <strong>of</strong> breast<br />

cancer. Ann Epidemiol 1997;7(6):375-82.<br />

16. Frisch RE, Wyshak G, Albright NL, Albright TE, Schiff I, Jones KP, et al.<br />

Lower prevalence <strong>of</strong> breast cancer and cancers <strong>of</strong> the reproductive system among<br />

former college athletes compared to non-athletes. Br J Cancer 1985;52(6):885-91.<br />

17. Luoto R, Latikka P, Pukkala E, Hakulinen T, Vihko V. The effect <strong>of</strong> physical<br />

activity on breast cancer risk: a cohort study <strong>of</strong> 30,548 women. Eur J Epidemiol<br />

2000;16(10):973-80.<br />

18. McTiernan A, Kooperberg C, White E, Wilcox S, Coates R, Adams-Campbell LL,<br />

et al. Recreational physical activity and the risk <strong>of</strong> breast cancer in postmenopausal<br />

women: the Women's Health Initiative Cohort Study. JAMA 2003;290(10):1331-6.<br />

19. Moore DB, Folsom AR, Mink PJ, Hong CP, Anderson KE, Kushi LH.<br />

Physical activity and incidence <strong>of</strong> postmenopausal breast cancer. Epidemiology<br />

2000;11(3):292-6.<br />

20. Moradi T, Nyren O, Zack M, Magnusson C, Persson I, Adami HO. Breast<br />

cancer risk and lifetime leisure-time and occupational physical activity (Sweden).<br />

Cancer Causes Control 2000;11(6):523-31.<br />

21. Paffenbarger RS, Jr., Lee IM, Wing AL. The influence <strong>of</strong> physical activity<br />

on the incidence <strong>of</strong> site-specific cancers in college alumni. Adv Exp Med Biol<br />

1992;322:7-15.<br />

22. Pukkala E, Poskiparta M, Apter D, Vihko V. Life-long physical activity and<br />

cancer risk among Finnish female teachers. Eur J Cancer Prev 1993;2(5):369-76.<br />

23. Rockhill B, Willett WC, Hunter DJ, Manson JE, Hankinson SE, Colditz GA.<br />

A prospective study <strong>of</strong> recreational physical activity and breast cancer risk. Arch<br />

Intern Med 1999;159(19):2290-6.<br />

24. Rockhill B, Willett WC, Hunter DJ, Manson JE, Hankinson SE, Spiegelman<br />

D, et al. Physical activity and breast cancer risk in a cohort <strong>of</strong> young women. J<br />

Natl Cancer Inst 1998;90(15):1155-60.<br />

25. Sesso HD, Paffenbarger RS, Jr., Lee IM. Physical activity and breast cancer risk in the<br />

College Alumni Health Study (United States). Cancer Causes Control 1998;9(4):433-9.<br />

26. Steenland K, Nowlin S, Palu S. Cancer incidence in the National Health and<br />

Nutrition Survey I. Follow-up data: diabetes, cholesterol, pulse and physical<br />

activity. Cancer Epidemiol Biomarkers Prev 1995;4(8):807-11.<br />

27. Thune I, Brenn T, Lund E, Gaard M. Physical activity and the risk <strong>of</strong> breast<br />

cancer. N Engl J Med 1997;336(18):1269-75.<br />

28.Vena JE, Graham S, Zielezny M, Brasure J, Swanson MK. Occupational exercise<br />

and risk <strong>of</strong> cancer. Am J Clin Nutr 1987;45(1 Suppl):318-27.<br />

29. Wyshak G, Frisch RE. Breast cancer among former college athletes compared<br />

to non-athletes: a 15-year follow-up. Br J Cancer 2000;82(3):726-30.<br />

30. Zheng W, Shu XO, McLaughlin JK, Chow WH, Gao YT, Blot WJ. Occupational<br />

physical activity and the incidence <strong>of</strong> cancer <strong>of</strong> the breast, corpus uteri, and ovary<br />

in Shanghai. Cancer 1993;71(11):3620-4.<br />

31. Lee IM, Cook NR, Rexrode KM, Buring JE. Lifetime physical activity and risk<br />

<strong>of</strong> breast cancer. Br J Cancer 2001;85(7):962-5.<br />

32. Gilliland FD, Li YF, Baumgartner K, Crumley D, Samet JM. Physical activity<br />

and breast cancer risk in hispanic and non-hispanic white women. Am J Epidemiol<br />

2001;154(5):442-50.<br />

33. Hirose K, Tajima K, Hamajima N, Inoue M, Takezaki T, Kuroishi T, et al. A<br />

large-scale, hospital-based case-control study <strong>of</strong> risk factors <strong>of</strong> breast cancer<br />

according to menopausal status. Jpn J Cancer Res 1995;86(2):146-54.<br />

34. Hu YH, Nagata C, Shimizu H, Kaneda N, Kashiki Y. <strong>Association</strong> <strong>of</strong> body<br />

mass index, physical activity, and reproductive histories with breast cancer: a<br />

case-control study in Gifu, Japan. Breast Cancer Res Treat 1997;43(1):65-72.<br />

35. Slattery ML, Edwards S, Murtaugh MA, Sweeney C, Herrick J, Byers T, et<br />

al. Physical Activity and Breast Cancer Risk among Women in the Southwestern<br />

United States. Ann Epidemiol 2007;17(5):342-353.<br />

36. Ueji M, Ueno E, Osei-Hyiaman D, Takahashi H, Kano K. Physical activity and<br />

the risk <strong>of</strong> breast cancer: a case-control study <strong>of</strong> Japanese women. J Epidemiol<br />

1998;8(2):116-22.<br />

37. Bernstein L, Henderson BE, Hanisch R, Sullivan-Halley J, Ross RK. Physical<br />

exercise and reduced risk <strong>of</strong> breast cancer in young women. J Natl Cancer Inst<br />

1994;86(18):1403-8.<br />

38. Friedenreich CM, Rohan TE. Physical activity and risk <strong>of</strong> breast cancer. Eur<br />

J Cancer Prev 1995;4(2):145-51.<br />

39. Verloop J, Rookus MA, van der Kooy K, van Leeuwen FE. Physical activity and<br />

breast cancer risk in women aged 20-54 years. J Natl Cancer Inst 2000;92(2):128-35.<br />

40. Coogan PF, Clapp RW, Newcomb PA, Mittendorf R, Bogdan G, Baron<br />

JA, et al. Variation in female breast cancer risk by occupation. Am J Ind Med<br />

1996;30(4):430-7.<br />

41. McTiernan A, Stanford JL, Weiss NS, Daling JR, Voigt LF. Occurrence <strong>of</strong><br />

breast cancer in relation to recreational exercise in women age 50-64 years.<br />

Epidemiology 1996;7(6):598-604.<br />

42. D'Avanzo B, Nanni O, La Vecchia C, Franceschi S, Negri E, Giacosa A, et al. Physical<br />

activity and breast cancer risk. Cancer Epidemiol Biomarkers Prev 1996;5(3):155-60.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 41


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

43. Levi F, Pasche C, Lucchini F, La Vecchia C. Occupational and leisure time<br />

physical activity and the risk <strong>of</strong> breast cancer. Eur J Cancer 1999;35(5):775-8.<br />

44. Mezzetti M, La Vecchia C, Decarli A, Boyle P, Talamini R, Franceschi S.<br />

Population attributable risk for breast cancer: diet, nutrition, and physical exercise.<br />

J Natl Cancer Inst 1998;90(5):389-94.<br />

45. Hulka BS. Epidemiology <strong>of</strong> susceptibility to breast cancer. Prog Clin Biol<br />

Res 1996;395:159-74.<br />

46. The Endogenous Hormones and Breast Cancer Collaborative Group.<br />

Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis<br />

<strong>of</strong> nine prospective studies. J Natl Cancer Inst 2002;94(8):606-16.<br />

47. Frisch RE, Gotz-Welbergen AV, McArthur JW, Albright T, Witschi J, Bullen<br />

B, et al. Delayed menarche and amenorrhea <strong>of</strong> college athletes in relation to age<br />

<strong>of</strong> onset <strong>of</strong> training. JAMA 1981;246(14):1559-63.<br />

48. Frisch RE, Wyshak G, Vincent L. Delayed menarche and amenorrhea in ballet<br />

dancers. N Engl J Med 1980;303(1):17-9.<br />

49. Bernstein L, Ross RK, Lobo RA, Hanisch R, Krailo MD, Henderson BE. The<br />

effects <strong>of</strong> moderate physical activity on menstrual cycle patterns in adolescence:<br />

implications for breast cancer prevention. Br J Cancer 1987;55(6):681-5.<br />

50. Bonen A, Ling WY, MacIntyre KP, Neil R, McGrail JC, Belcastro AN. Effects<br />

<strong>of</strong> exercise on the serum concentrations <strong>of</strong> FSH, LH, progesterone, and estradiol.<br />

Eur J Appl Physiol Occup Physiol 1979;42(1):15-23.<br />

51. Bullen BA, Skrinar GS, Beitins IZ, von Mering G, Turnbull BA, McArthur JW.<br />

Induction <strong>of</strong> menstrual disorders by strenuous exercise in untrained women. N<br />

Engl J Med 1985;312(21):1349-53.<br />

52. Siiteri PK. Adipose tissue as a source <strong>of</strong> hormones. Am J Clin Nutr 1987;45(1<br />

Suppl):277-82.<br />

53.Cauley JA, Gutai JP, Kuller LH, LeDonne D, Powell JG. The epidemiology <strong>of</strong> serum<br />

sex hormones in postmenopausal women. Am J Epidemiol 1989;129(6):1120-31.<br />

54. Nelson ME, Meredith CN, Dawson-Hughes B, Evans WJ. Hormone and bone<br />

mineral status in endurance-trained and sedentary postmenopausal women. J Clin<br />

Endocrinol Metab 1988;66(5):927-33.<br />

55. Verkasalo PK, Thomas HV, Appleby PN, Davey GK, Key TJ. Circulating<br />

levels <strong>of</strong> sex hormones and their relation to risk factors for breast cancer: a<br />

cross-sectional study in 1092 pre- and postmenopausal women (United Kingdom).<br />

Cancer Causes Control 2001;12(1):47-59.<br />

56. Newcomb PA, Klein R, Klein BE, Haffner S, Mares-Perlman J, Cruickshanks<br />

KJ, et al. <strong>Association</strong> <strong>of</strong> dietary and life-style factors with sex hormones in<br />

postmenopausal women. Epidemiology 1995;6(3):318-21.<br />

57. McTiernan A, Tworoger S, Schwartz RS, Ulrich CM, Yasui Y, Irwin ML, et al.<br />

Effect <strong>of</strong> Exercise on Serum Estrogen in Postmenopausal Women: a 12-Month<br />

Randomized Controlled Trial. Cancer Research 2004;64(8):2923-2928.<br />

58. McTiernan A, Tworoger S, Rajan B, Yasui Y, Sorenson B, Ulrich C, et al. Effect<br />

<strong>of</strong> exercise on serum androgens in postmenopausal women: a 12-month randomized<br />

clinical trial. Cancer Epidemiology, Biomarkers & Prevention 2004;13(7):1-7.<br />

59. McTiernan A, Wu L, Chen C, Chlebowski R, Mossavar-Rahmani Y, Modugno<br />

F, et al. Relation <strong>of</strong> BMI and physical activity to sex hormones in postmenopausal<br />

women. Obesity (Silver Spring) 2006;14(9):1662-77.<br />

60. Flegal KM, Carroll MD, Kuczmarski RJ, Johnson CL. Overweight and obesity<br />

in the United States: prevalence and trends, 1960-1994. Int J Obes Relat Metab<br />

Disord 1998;22(1):39-47.<br />

61. Friedenreich CM. Review <strong>of</strong> anthropometric factors and breast cancer risk.<br />

Eur J Cancer Prev 2001;10(1):15-32.<br />

62. Women's Health Initiative Study Group. Design <strong>of</strong> the Women's Health Initiative<br />

clinical trial and observational study. Control Clin Trials 1998;19(1):61-109.<br />

63. Morimoto LM, White E, Chen Z, Chlebowski RT, Hays J, Kuller L, et al.<br />

Obesity, body size, and risk <strong>of</strong> postmenopausal breast cancer: the Women's Health<br />

Initiative (United States). Cancer Causes Control 2002;13(8):741-51.<br />

64. Huang Z, Hankinson SE, Colditz GA, Stampfer MJ, Hunter DJ, Manson<br />

JE, et al. Dual effects <strong>of</strong> weight and weight gain on breast cancer risk. JAMA<br />

1997;278(17):1407-11.<br />

65. Renehan AG TM, Egger M, Heller RF, Zwahlen M. Body-mass index and<br />

incidence <strong>of</strong> cancer: a systematic review and meta-analysis <strong>of</strong> prospective<br />

observational studies. Lancet 2008;371(9612):569-78.<br />

66. Michels KB Terry KL, Willett WC. Longitudinal study on the role <strong>of</strong> body size<br />

in premenopausal breast cancer. Arch Intern Med 2006 Nov 27;166(21):2395-402.<br />

67. El Saghir NS KM, Eid T, El Kinge AR, Charafeddine M, Geara F, Seoud M,<br />

Shamseddine AI. Trends in epidemiology and management <strong>of</strong> breast cancer in<br />

developing <strong>Arab</strong> countries: a literature and registry analysis. Int J Surg 2007<br />

Aug;5(4):225-33.<br />

68. Metwally M LT, Ledger WL. The impact <strong>of</strong> obesity on female reproductive<br />

function. Obes Rev 2007 Nov;8(6):515-23.<br />

69. Ballard-Barbash R, Schatzkin A, Taylor PR, Kahle LL. <strong>Association</strong> <strong>of</strong> change<br />

in body mass with breast cancer. Cancer Res 1990;50(7):2152-5.<br />

70. Brinton LA, Swanson CA. Height and weight at various ages and risk <strong>of</strong> breast<br />

cancer. Ann Epidemiol 1992;2(5):597-609.<br />

71. Enger SM, Ross RK, Paganini-Hill A, Carpenter CL, Bernstein L. Body size,<br />

physical activity, and breast cancer hormone receptor status: results from two<br />

case-control studies. Cancer Epidemiol Biomarkers Prev 2000;9(7):681-7.<br />

72. Folsom AR, Kaye SA, Prineas RJ, Potter JD, Gapstur SM, Wallace RB.<br />

Increased incidence <strong>of</strong> carcinoma <strong>of</strong> the breast associated with abdominal adiposity<br />

in postmenopausal women. Am J Epidemiol 1990;131(5):794-803.<br />

73. Lubin F, Ruder AM, Wax Y, Modan B. Overweight and changes in weight<br />

throughout adult life in breast cancer etiology. A case-control study. Am J Epidemiol<br />

1985;122(4):579-88.<br />

74. Magnusson C, Baron J, Persson I, Wolk A, Bergstrom R, Trichopoulos D, et<br />

al. Body size in different periods <strong>of</strong> life and breast cancer risk in post-menopausal<br />

women. Int J Cancer 1998;76(1):29-34.<br />

75. Radimer K, Siskind V, Bain C, Sch<strong>of</strong>ield F. Relation between anthropometric<br />

indicators and risk <strong>of</strong> breast cancer among Australian women. Am J Epidemiol<br />

1993;138(2):77-89.<br />

76. Ziegler RG, Hoover RN, Nomura AM, West DW, Wu AH, Pike MC, et al.<br />

Relative weight, weight change, height, and breast cancer risk in Asian-American<br />

women. J Natl Cancer Inst 1996;88(10):650-60.<br />

77. Kaaks R, Van Noord PA, Den Tonkelaar I, Peeters PJ, Riboli E, Grobbee DE.<br />

Breast-cancer incidence in relation to height, weight and body-fat distribution in<br />

the Dutch "DOM" cohort. Int J Cancer 1998;76(5):647-51.<br />

78. Sellers TA, Kushi LH, Potter JD, Kaye SA, Nelson CL, McGovern PG, et al.<br />

Effect <strong>of</strong> family history, body-fat distribution, and reproductive factors on the risk<br />

<strong>of</strong> postmenopausal breast cancer. N Engl J Med 1992;326(20):1323-9.<br />

79. Trentham-Dietz A, Newcomb PA, Storer BE, Longnecker MP, Baron J, Greenberg<br />

ER, et al. Body size and risk <strong>of</strong> breast cancer. Am J Epidemiol 1997;145(11):1011-9.<br />

80. Coates RJ, Uhler RJ, Hall HI, Potischman N, Brinton LA, Ballard-Barbash<br />

R, et al. Risk <strong>of</strong> breast cancer in young women in relation to body size and weight<br />

gain in adolescence and early adulthood. Br J Cancer 1999;81(1):167-74.<br />

81. Eliassen AH, Colditz GA, Rosner B, Willett WC, Hankinson SE. Adult weight<br />

change and risk <strong>of</strong> postmenopausal breast cancer. JAMA 2006;296(2):193-201.<br />

82. Corbould AM, Judd SJ, Rodgers RJ. Expression <strong>of</strong> types 1, 2, and 3 17 betahydroxysteroid<br />

dehydrogenase in subcutaneous abdominal and intra-abdominal<br />

adipose tissue <strong>of</strong> women. J Clin Endocrinol Metab 1998;83(1):187-94.<br />

83. McTiernan A, Ulrich C, Slate S, Potter J. Physical activity and cancer etiology:<br />

associations and mechanisms. Cancer Causes Control 1998;9(5):487-509.<br />

84. Hankinson SE, Willett WC, Colditz GA, Hunter DJ, Michaud DS, Deroo B, et<br />

42 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


al. Circulating concentrations <strong>of</strong> insulin-like growth factor-I and risk <strong>of</strong> breast<br />

cancer. Lancet 1998;351(9113):1393-6.<br />

85. McCarty MF. Up-regulation <strong>of</strong> IGF binding protein-1 as an anticarcinogenic<br />

strategy: relevance to caloric restriction, exercise, and insulin sensitivity. Med<br />

Hypotheses 1997;48(4):297-308.<br />

86. Schmitz KH, Ahmed RL, Yee D. Effects <strong>of</strong> a nine-month strength training<br />

intervention on insulin, IGF-1, IGFBP1, and IGFBP-3 in 30-50 year old women.<br />

Cancer Epidemiol Biomarkers Prev 2002;11:1597-1604.<br />

87. Marshall JR, Qu Y, Chen J, Parpia B, Campbell TC. Additional ecological<br />

evidence: lipids and breast cancer mortality among women aged 55 and over in<br />

China. Eur J Cancer 1992;28A(10):1720-7.<br />

88. Prentice RL, Kakar F, Hursting S, Sheppard L, Klein R, Kushi LH. Aspects <strong>of</strong><br />

the rationale for the Women's Health Trial. J Natl Cancer Inst 1988;80(11):802-14.<br />

89. Welsch CW. Relationship between dietary fat and experimental mammary<br />

tumorigenesis: a review and critique. Cancer Res 1992;52(7 Suppl):2040S-2048S.<br />

90. Thompson HJ ZZ, Jiang W. Weight control and breast cancer prevention:<br />

are the effects <strong>of</strong> reduced energy intake equivalent to those <strong>of</strong> increased energy<br />

expenditure? J Nutr 2004 Dec;134(12 Suppl):3407S-3411S.<br />

91. Zhu Z JW, Sells JL, Neil ES, McGinley JN, Thompson HJ. Effect <strong>of</strong> nonmotorized<br />

wheel running on mammary carcinogenesis: circulating biomarkers, cellular<br />

processes, and molecular mechanisms in rats. Cancer Epidemiol Biomarkers<br />

Prev 2008 Aug;17(8):1920-9.<br />

92. Boyd NF, Greenberg C, Lockwood G, Little L, Martin L, Byng J, et al. Effects at<br />

two years <strong>of</strong> a low-fat, high-carbohydrate diet on radiologic features <strong>of</strong> the breast:<br />

results from a randomized trial. Canadian Diet and Breast Cancer Prevention<br />

Study Group. J Natl Cancer Inst 1997;89(7):488-96.<br />

93. Wu AH, Ziegler RG, Nomura AM, West DW, Kolonel LN, Horn-Ross PL, et al.<br />

Soy intake and risk <strong>of</strong> breast cancer in Asians and Asian Americans. Am J Clin<br />

Nutr 1998;68(6 Suppl):1437S-1443S.<br />

94. Holmes MD, Schisterman EF, Spiegelman D, Hunter DJ, Willett WC. Re:<br />

Meta-analysis: dietary fat intake, serum estrogen levels, and the risk <strong>of</strong> breast<br />

cancer. J Natl Cancer Inst 1999;91(17):1511-2.<br />

95. Prentice RL, Caan B, Chlebowski RT, Patterson R, Kuller LH, Ockene JK,<br />

et al. Low-fat dietary pattern and risk <strong>of</strong> invasive breast cancer: the Women's<br />

Health Initiative Randomized Controlled Dietary Modification Trial. Jama<br />

2006;295(6):629-42.<br />

96. Smith-Warner SA, Spiegelman D, Yaun SS, Adami HO, Beeson WL, van den<br />

Brandt PA, et al. Intake <strong>of</strong> fruits and vegetables and risk <strong>of</strong> breast cancer: a pooled<br />

analysis <strong>of</strong> cohort studies. JAMA 2001;285(6):769-76.<br />

97. Pillow PC, Duphorne CM, Chang S, Contois JH, Strom SS, Spitz MR, et<br />

al. Development <strong>of</strong> a database for assessing dietary phytoestrogen intake. Nutr<br />

Cancer 1999;33(1):3-19.<br />

98. Jones JL, Daley BJ, Enderson BL, Zhou JR, Karlstad MD. Genistein inhibits<br />

tamoxifen effects on cell proliferation and cell cycle arrest in T47D breast cancer<br />

cells. Am Surg 2002;68(6):575-7; discussion 577-8.<br />

99. Ju YH, Doerge DR, Allred KF, Allred CD, Helferich WG. Dietary genistein<br />

negates the inhibitory effect <strong>of</strong> tamoxifen on growth <strong>of</strong> estrogen-dependent<br />

human breast cancer (MCF-7) cells implanted in athymic mice. Cancer Res<br />

2002;62(9):2474-7.<br />

100. Adlercreutz CH, Goldin BR, Gorbach SL, Hockerstedt KA, Watanabe S,<br />

Hamalainen EK, et al. Soybean phytoestrogen intake and cancer risk. J Nutr<br />

1995;125(3 Suppl):757S-770S.<br />

101. Adlercreutz H, Hamalainen E, Gorbach S, Goldin B. Dietary phyto-oestrogens<br />

and the menopause in Japan. Lancet 1992;339(8803):1233.<br />

102. Cassidy A, Bingham S, Carlson J, et al. Biological effects <strong>of</strong> plant oestrogens<br />

in premenopausal women. FASEB 1993;7:A866.<br />

103. Shin MH, Holmes MD, Hankinson SE, Wu K, Colditz GA, Willett WC. Intake<br />

<strong>of</strong> dairy products, calcium, and vitamin d and risk <strong>of</strong> breast cancer. J Natl Cancer<br />

Inst 2002;94(17):1301-11.<br />

104. Dorgan JF, Sowell A, Swanson CA, Potischman N, Miller R, Schussler N, et<br />

al. Relationships <strong>of</strong> serum carotenoids, retinol, alpha-tocopherol, and selenium<br />

with breast cancer risk: results from a prospective study in Columbia, Missouri<br />

(United States). Cancer Causes Control 1998;9(1):89-97.<br />

105. Hulten K, Van Kappel AL, Winkvist A, Kaaks R, Hallmans G, Lenner P, et<br />

al. Carotenoids, alpha-tocopherols, and retinol in plasma and breast cancer risk<br />

in northern Sweden. Cancer Causes Control 2001;12(6):529-37.<br />

106. Sato R, Helzlsouer KJ, Alberg AJ, H<strong>of</strong>fman SC, Norkus EP, Comstock GW.<br />

Prospective study <strong>of</strong> carotenoids, tocopherols, and retinoid concentrations and<br />

the risk <strong>of</strong> breast cancer. Cancer Epidemiol Biomarkers Prev 2002;11(5):451-7.<br />

107. Cui Y RT. Vitamin D, calcium, and breast cancer risk: a review. Cancer<br />

Epidemiol Biomarkers Prev 2006 Aug;15(8):1427-37.<br />

108. Al-Nozha MM A-MY, Al-Maatouq MA, Arafah MR, Khalil MZ, Khan NB,<br />

Al-Marzouki K, Abdullah MA, Al-Khadra AH, Al-Harthi SS, Al-Shahid MS, Al-<br />

Mobeireek A, Nouh MS. Obesity in Saudi <strong>Arab</strong>ia. Saudi Med J 2005 May;26(5):824-9.<br />

109. Al-Nozha MM A-HH, Arafah MR, Al-Khadra A, Al-Mazrou YY, Al-Maatouq<br />

MA, Khan NB, Al-Marzouki K, Al-Harthi SS, Abdullah M, Al-Shahid MS.<br />

Prevalence <strong>of</strong> physical activity and inactivity among Saudis aged 30-70 years.<br />

A population-based cross-sectional study. Saudi Med J 2007 Apr;28(4):559-68.<br />

110. Ghannam NN HM, Bakheet SM, Khan BA. Bone mineral density <strong>of</strong> the spine<br />

and femur in healthy Saudi females: relation to vitamin D status, pregnancy, and<br />

lactation. Calcif Tissue Int 1999;65(1):23-8.<br />

111. Chlebowski RT, Aiello E, McTiernan A. Weight loss in breast cancer patient<br />

management. J Clin Oncol 2002;20(4):1128-43.<br />

112. Brown JK, Byers T, Doyle C, Coumeya KS, Demark-Wahnefried W, Kushi<br />

LH, et al. Nutrition and physical activity during and after cancer treatment:<br />

an American Cancer Society guide for informed choices. CA Cancer J Clin<br />

2003;53(5):268-91.<br />

113. Folsom A, Kaye S, Sellers T, Hong C-P, Cerhan J, Potter J, et al. Body fat<br />

distribution and 5-year risk <strong>of</strong> death in older women. JAMA 1993;269(4):483-87.<br />

114. Dignam JJ, Wieand K, Johnson KA, Fisher B, Xu L, Mamounas EP. Obesity,<br />

tamoxifen use, and outcomes in women with estrogen receptor-positive early-stage<br />

breast cancer. J Natl Cancer Inst 2003;95(19):1467-76.<br />

115. Berclaz G, Li S, Price K, Coates A, Castiglione-Gertsch M, Rudenstam<br />

C-M, et al. Body mass index as a prognostic feature in I operable breast cancer:<br />

the International Breast Cancer Study Group experience. Ann Oncol 15:875-84.<br />

116. Goodwin PJ, Esplen MJ, Winocur J, Butler K, Pritchard KI. Development<br />

<strong>of</strong> a weight management program in women with newly diagnosed locoregional<br />

breast cancer. In: Bitzer J, Stauber M, editors. Psychosomatic Obstetrics and<br />

Gynecology. Bologna (Italy): Monduzzi Editore, International Proceedings<br />

Division; 1995. p. 491-6.<br />

117. Chang S, Alderfer J, Asmar L, Buzdar A. Inflammatory breast cancer survival:<br />

the role <strong>of</strong> obesity and menopausal status at diagnosis. . Breast Cancer Res Treat<br />

2001;64(2):157-63.<br />

118. Goodwin PJ, Ennis M, Pritchard KI, Trudeau ME, Koo J, Madarnas Y, et al.<br />

Fasting insulin and outcome in early-stage breast cancer: results <strong>of</strong> a prospective<br />

cohort study. J Clin Oncol 2002;20(1):42-51.<br />

119. Borugian M, Sheps S, Kim-Sing C, Olivotto I, Van Patten C, Dunn B, et al.<br />

Waist-to-hip ratio and breast cancer mortality. Am J Epidemiol 2003;158(10):963-8.<br />

120. Kumar N, Cantor A, Allen K, Cox C. Android obesity at diagnosis and breast<br />

carcinoma survival: Evaluation <strong>of</strong> the effects <strong>of</strong> anthropometric variables at<br />

diagnosis, including body composition and body fat distribution and weight gain<br />

during life span, and survival from breast carcinoma. Cancer 2000;88(12):2751-7.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 43


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

121. Li CI, Malone KE, Porter PL, Daling JR. Epidemiologic and molecular risk factors<br />

for contralateral breast cancer among young women. Br J Cancer 2003;89(3):513-8.<br />

122.Goodwin PJ, Ennis M, Pritchard KI, McCready D, Koo J, Sidl<strong>of</strong>sky S, et<br />

al. Adjuvant treatment and onset <strong>of</strong> menopause predict weight gain after breast<br />

cancer diagnosis. J Clin Oncol 1999;17(1):120-9.<br />

123. Irwin ML, Crumley D, McTiernan A, Bernstein L, Baumgartner R, Gilliland FD,<br />

et al. Physical activity levels before and after a diagnosis <strong>of</strong> breast carcinoma: the<br />

Health, Eating, Activity, and Lifestyle (HEAL) study. Cancer 2003;97(7):1746-57.<br />

124. Rock C, McEligot A, Flatt S. Eating pathology and obesity in women at risk<br />

for breast cancer recurrence. Int J Eat Disord 2000;27(2):172-9.<br />

125. Fisher B, Costantino JP, Wickerham DL, Redmond CK, Kavanah M, Cronin<br />

WM, et al. Tamoxifen for prevention <strong>of</strong> breast cancer: report <strong>of</strong> the National<br />

Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst<br />

1998;90(18):1371-88.<br />

126. Demark-Wahnefried W, Winer E, Rimer B. Why women gain weight with adjuvant<br />

chemotherapy for breast cancer. (review). J Clin Oncol 1993;11(7):1418-29.<br />

127. Shepherd L, Parulekar W, Day A. Weight gain during adjuvant<br />

therapy in high risk pre/perimenopausal breast cancer patients: analysis<br />

<strong>of</strong> a National Cancer Institute <strong>of</strong> Canada Clinical Trials Group (NCIC<br />

CTG) Phase III Study. Proc Amer Soc Clin Oncol 2001;20(36a).<br />

128. Bernstein L, Deapen D, Cerhan JR, Schwartz SM, Liff J, McGann-Maloney<br />

E, et al. Tamoxifen therapy for breast cancer and endometrial cancer risk. J Natl<br />

Cancer Inst 1999;91(19):1654-62.<br />

129. Ganz PA, Rowland JH, Meyerowitz BE, Desmond KA. Impact <strong>of</strong> different<br />

adjuvant therapy strategies on quality <strong>of</strong> life in breast cancer survivors. Recent<br />

Results Cancer Res 1998;152:396-411.<br />

130. Courneya KS. Exercise in cancer survivors: an overview <strong>of</strong> research. Med<br />

Sci Sports Exerc 2003;35(11):1846-52.<br />

131. Holmes MD, Chen WY, Feskanich D, Kroenke CH, Colditz GA. Physical<br />

activity and survival after breast cancer diagnosis. JAMA 2005;293(20):2479-86.<br />

132. Abrahamson PE, Gammon MD, Lund MJ, Britton JA, Marshall SW, E.W. F,<br />

et al. Recreational physical activity and survival among young women with breast<br />

cancer. Cancer 2006;107(8):1777-85.<br />

133. Chlebowski RT, Blackburn GL, Thomson CA, Nixon DW, Shapiro A, Hoy MK,<br />

et al. Dietary fat reduction and breast cancer outcome: interim efficacy results from<br />

the Women's Intervention Nutrition Study. J Natl Cancer Inst 2006;98(24):1767-76.<br />

134. Howell A, on behalf <strong>of</strong> the ATAC Trialists' Group A. The ATAC ('Arimidex',<br />

Tamoxifen, Alone or in Combination) trial in postmenopausal women with early<br />

breast cancer - updated efficacy results based on a median follow-up <strong>of</strong> 5 years.<br />

Breast Cancer Research and Treatment 2004;88(1 Suppl).<br />

135. Chlebowski RT, Col N, Winer EP, Collyar DE, Cummings SR, Vogel VG, 3rd, et<br />

al. American Society <strong>of</strong> Clinical <strong>Oncology</strong> technology assessment <strong>of</strong> pharmacologic<br />

interventions for breast cancer risk reduction including tamoxifen, raloxifene, and<br />

aromatase inhibition. J Clin Oncol 2002;20(15):3328-43.<br />

136. McTiernan A, Rajan KB, Tworoger SS, Irwin M, Bernstein L, Baumgartner<br />

R, et al. Adiposity and sex hormones in postmenopausal breast cancer survivors.<br />

J Clin Oncol 2003;21(10):1961-6.<br />

137. Calle E, Kaaks R. Overweight, obesity and cancer: epidemiological evidence<br />

and proposed mechanisms. Nat Rev Cancer 2004;4(8):579-91.<br />

138. Irwin ML, Yasui Y, Ulrich CM, Bowen D, Rudolph RE, Schwartz RS, et al.<br />

Effect <strong>of</strong> exercise on total and intra-abdominal body fat in postmenopausal women:<br />

a randomized controlled trial. JAMA 2003;289(3):323-30.<br />

139. McTiernan A, Ulrich C, Kumai C, Bean D, Schwartz R, Mahloch J, et al.<br />

Anthropometric and hormone effects <strong>of</strong> an eight-week exercise-diet intervention<br />

in breast cancer patients: results <strong>of</strong> a pilot study. Cancer Epidemiol Biomarkers<br />

Prev 1998;7(6):477-81.<br />

140. Nieman DC, Cook VD, Henson DA, Suttles J, Rejeski WJ, Ribisl PM, et al.<br />

Moderate exercise training and natural killer cell cytotoxic activity in breast<br />

cancer patients. Int J Sports Med 1995;16(5):334-7.<br />

141. Obesity Education Initiative. Clinical Guidelines on the Identification,<br />

Evaluation, and Treatment <strong>of</strong> Overweight and Obesity in Adults: The Evidence Report,<br />

Executive Summary. Bethesda, MD: National Heart, Lung, and Blood Institute; 1998.<br />

142. Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker<br />

EA, et al. Reduction in the incidence <strong>of</strong> type 2 diabetes with lifestyle intervention<br />

or metformin. N Engl J Med 2002;346(6):393-403.<br />

143. McTigue KM, Harris R, Hemphill B, Lux L, Sutton S, Bunton AJ, et al.<br />

Screening and interventions for obesity in adults: summary <strong>of</strong> the evidence for<br />

the U.S. Preventive Services Task Force. Ann Intern Med 2003;139(11):933-49.<br />

144. Samaha FF, Iqbal N, Seshadri P, Chicano KL, Daily DA, McGrory J, et al.<br />

A low-carbohydrate as compared with a low-fat diet in severe obesity. N Engl J<br />

Med 2003;348(21):2074-81.<br />

145. Foster GD, Wyatt HR, Hill JO, McGuckin BG, Brill C, Mohammed BS,<br />

et al. A randomized trial <strong>of</strong> a low-carbohydrate diet for obesity. N Engl J Med<br />

2003;348(21):2082-90.<br />

146. Arterburn D, Noel P. Effects <strong>of</strong> drug treatment for obesity in adults. Clin<br />

Evid 2001:412-19.<br />

147. Brolin RE. Bariatric surgery and long-term control <strong>of</strong> morbid obesity. Jama<br />

2002;288(22):2793-6.<br />

148. Pronk NP, Wing RR. Physical activity and long-term maintenance <strong>of</strong> weight<br />

loss. Obes Res 1994;2:587-99.<br />

149. Wilmore JH. Increasing physical activity: alterations in body mass and<br />

composition. Am J Clin Nutr 1996;63(3 Suppl):456S-460S.<br />

150. Loprinzi CL, Athmann LM, Kardinal CG, O'Fallon JR, See JA, Bruce BK, et<br />

al. Randomized trial <strong>of</strong> dietician counseling to try to prevent weight gain associated<br />

with breast cancer adjuvant chemotherapy. <strong>Oncology</strong> 1996;53(3):228-32.<br />

151. Djuric Z, DiLaura NM, Jenkins I, Darga L, Jen CK, Mood D, et al. Combining<br />

weight-loss counseling with the weight watchers plan for obese breast cancer<br />

survivors. Obes Res 2002;10(7):657-65.<br />

152. Jen K, Djuric Z, DiLaura N, Buison A, Redd J, Maranci V, et al. Improvement<br />

<strong>of</strong> metabolism among obese breast cancer survivors in differing weight loss<br />

regimens. Obes Res 2004;12(2):306-12.<br />

153. de Waard F, Ramlau R, Mulders Y, de Vries T, van Waveren S. A feasibility<br />

study on weight reduction in obese postmenopausal breast cancer patients. Eur<br />

J Cancer Prev 1993;2(3):233-8.<br />

154. Mefferd K, Nichols JF, Pakiz B, Rock CL. A cognitive behavioral therapy<br />

intervention to promote weight loss improves body composition and blood lipid<br />

pr<strong>of</strong>iles among overweight breast cancer survivors. Breast Cancer Res Treat<br />

2006;[Epub ahead <strong>of</strong> print].<br />

155. Chlebowski RT, Blackburn GL, Buzzard IM, Rose DP, Martino S, Khandekar<br />

JD, et al. Adherence to a dietary fat intake reduction program in postmenopausal<br />

women receiving therapy for early breast cancer. The Women's Intervention<br />

Nutrition Study. J Clin Oncol 1993;11(11):2072-80.<br />

156.Pierce JP, Faerber S, Wright FA, Newman V, Flatt SW, Kealey S, et al.<br />

Feasibility <strong>of</strong> a randomized trial <strong>of</strong> a high-vegetable diet to prevent breast cancer<br />

recurrence. Nutr Cancer 1997;28(3):282-8.<br />

157.Chlebowski RT, Blackburn G, Winters B, Goodman M, et al. Long term<br />

adherence to dietary fat reduction in the Women's Intervention Nutrition Study.<br />

Proc Amer Soc Clin Oncol 2000;19:207.<br />

158. Thomson C, Rock C, Giuliano A, Newton T, Cui H, Reid P, et al. Longitudinal<br />

changes in body weight and body composition among women previously treated<br />

for breast cancer consuming a high-vegetable, fruit and fiber, low-fat diet. Eur<br />

J Nutr 2004;5:1-8.<br />

44 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


CHALLENGES OF CANCER SCREENING PROGRAM<br />

Abulkhair O. 1 , Al Amro A. 2 , Jazieh AR. 1<br />

(1) King Abdulaziz <strong>Medical</strong> City, Riyadh<br />

(2) King Fahad <strong>Medical</strong> City, Riyadh<br />

Corresponding Author: Dr. Omalkhair Abulkhair<br />

Section Head, Division <strong>of</strong> Adult <strong>Medical</strong> <strong>Oncology</strong><br />

Department <strong>of</strong> <strong>Oncology</strong>, King Abdulaziz <strong>Medical</strong> City<br />

P.O. Box 22490, Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

E-mail: abulkhairo@ngha.med.sa<br />

Background<br />

Cancer is a global public health problem. It is the second leading cause <strong>of</strong> death<br />

worldwide. It is <strong>of</strong>ten regarded as a disease cause <strong>of</strong> the developed world, but<br />

with improved living standards, incidence in low and middle income countries<br />

is on the rise. By the year 2030, seven out <strong>of</strong> every ten new cases will occur in<br />

the developing world.1<br />

Survival outcomes vary dramatically throughout the world and variation in access<br />

to quality cancer care is a major cause <strong>of</strong> these discrepancies.2 Over 40% <strong>of</strong> more<br />

than 7 million cancer deaths can be prevented. Furthermore, cancer is curable if<br />

detected early and treated adequately. This applies in particular to breast cancer,<br />

colon, prostate, and cervical cancer as the technology for screening, diagnosis<br />

and treating is mature.<br />

A recent publication about the future burden <strong>of</strong> breast cancer in Saudi <strong>Arab</strong>ia which<br />

anticipated the incidence and mortality <strong>of</strong> cases will increase by about 350% and<br />

160% respectively over a ten-year period.3<br />

Saudi Society for Cancer recognized the importance <strong>of</strong> prevention and early<br />

detection. In an effort to combat cancer through early detection, Abdulatif<br />

Charitable Screening Center was established as the first dedicated Cancer Screening<br />

Center in the Kingdom.<br />

The objective <strong>of</strong> this manuscript is to address the challenges/barriers which<br />

were encountered. Data related, center related, personal related challenges were<br />

identified, different interventions were implemented for each barrier. Furthermore,<br />

we will review the planning strategies for such project. This information may be <strong>of</strong><br />

benefit to health care providers, health care organizations and health care systems<br />

personnel when considering establishing public cancer screening programs.<br />

Readers are advised to review the World Health Organization (WHO) guide for<br />

effective programs which includes six modules that provide practical advice for<br />

program manager and policy makers on how to advocate, plan and implement<br />

effective cancer control programs, prevention and early detection.<br />

Methods<br />

The Abdulatiff Charitable Screening Center is the first center in Saudi <strong>Arab</strong>ia to<br />

conduct early diagnosis <strong>of</strong> breast cancer initially but then to cover screening for<br />

cervical, colon and prostate cancers. The Center inauguration has started screening<br />

in October 2007.<br />

The center is governed by Board <strong>of</strong> Directors which comprised <strong>of</strong> experts and<br />

leaders in the field <strong>of</strong> cancer diagnosis and treatment. The Center is empowered<br />

with a stand alone management and policy and procedure to ensure accurate<br />

screening and referral to tertiary care centers.<br />

Identifying Barriers<br />

The initial contact helped to identify centers barriers and concerns which can be<br />

summarized as follows:<br />

1. Concept Approval<br />

The involvement <strong>of</strong> charity organization in screening is <strong>of</strong> great concern since<br />

screening is a project which needs careful planning, budgeting, and control. The<br />

delay in establishing national screening program and cloudy strategy forced the<br />

Saudi Cancer Society to establish the first screening center. We realized that no<br />

public awareness program will be successful unless there are centers ready to<br />

screen candidates.<br />

2. Financial barriers<br />

Establishing screening center requires substantial financial support not only for<br />

the initial start up cost but for the running expenses <strong>of</strong> the center on ongoing<br />

basis especially if the center does not charge the patients. A decision was made<br />

to make screening free <strong>of</strong> charge in order to remove “out <strong>of</strong> pocket” expenses as<br />

barrier for participation.<br />

This center was a donation from a businessman who donated million Saudi Riyals<br />

which include the building and operation <strong>of</strong> first year. The plans took place to<br />

establish 8 million USD (30 million SR) endowment for society in Riyadh to<br />

secure permanent revenue to finance its activities.<br />

3. Personnel<br />

Assuring adequate staffing to run the center was major challenge which delayed<br />

the opening <strong>of</strong> the center for considerable time. There was a need for a family<br />

physician, a health educator, receptionist, mammography technician, and radiologist<br />

to read mammogram.<br />

4. Data Collection/Confidentiality:<br />

In order to identify possible risk factor and perform quality control and to develop<br />

a recall system, predetermined data should be collected and entered into unified<br />

database. This was an important barrier identified few months after operation<br />

and when number <strong>of</strong> cases increase and in order to overcome this problem, new<br />

s<strong>of</strong>tware will be applied.<br />

5. Recall System<br />

To overcome such problem, a form designed which has all personal information<br />

including ID and phone problem. Patient with abnormal mammogram findings<br />

where called back for further testing and work-up.<br />

6. Referral Process<br />

The center is designed to perform screening only. Therefore, collaboration with<br />

tertiary care centers was mandatory to ensure confirmation <strong>of</strong> diagnosis and<br />

further therapy. So, a proposal was submitted to all tertiary hospitals. However,<br />

for the initial 8 months, only two centers cooperate to have the suspicious cases<br />

for further investigation. So, lack <strong>of</strong> health facilities was an issue and we tried to<br />

overcome with cooperation with three tertiary care centers though, not all these<br />

hospitals have all needed resources in terms <strong>of</strong> performing investigation and<br />

access to cancer specialists.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 45


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

7. Barriers to Participation<br />

The center stared in August 2007, the flow <strong>of</strong> cases was very slow and that was<br />

attributed to cultural/social barriers that by diagnosis <strong>of</strong> breast cancer, women will<br />

lose her role in live.4 Lack <strong>of</strong> knowledge among female which has been reported<br />

by many studies done in different regions <strong>of</strong> Saudi <strong>Arab</strong>ia and concluded lack <strong>of</strong><br />

knowledge towards risk factors, breast cancer screening modality5, also diversity<br />

in health beliefs and behaviors which exist in religious subgroups.6 So, health<br />

communication should be modified to suit women in different groups to increase<br />

participation in screening.<br />

Other screening barriers include cultural knowledge and the use <strong>of</strong> traditional<br />

treatment,7 related fear, low self efficacy, fatalism, misinformation, and ineffective<br />

health communication.8 Through this center, more than 2500 ladies were screened;<br />

thus, far many participants barriers were identified. As listed above, the majority<br />

are related to attitude and lack <strong>of</strong> knowledge. To overcome this barrier, a wellorganized<br />

public health program was conducted throughout the year for ladies<br />

at work, schools, colleges/universities and public places such as Prince Salman<br />

Social Center, shopping mall, in addition to the use <strong>of</strong> media through a common<br />

popular TV program. This step had a good impact on increasing level <strong>of</strong> awareness<br />

and increase participation <strong>of</strong> women. This was noticed when the medial covered<br />

the opening <strong>of</strong> the center by USA former First Lady Laura Bush. The numbers<br />

<strong>of</strong> screened ladies increased 10 times over that before the opening ceremony,<br />

which reflects the importance and the vital role that media play in increasing<br />

public awareness. There was another important barrier that was reported through<br />

several studies which is the primary health care physician role in cancer prevention<br />

demonstrated by low adherence to prevention and screening recommendations as<br />

highlighted in several studies includes:9<br />

• Physician’s beliefs that prevention and not early detection <strong>of</strong> cancer is not<br />

part <strong>of</strong> their jobs.<br />

• Physicians / patients bond (lack <strong>of</strong> trust).<br />

• Lack <strong>of</strong> services and access to health care for prevention and screening.<br />

• Lack <strong>of</strong> time.<br />

• Lack <strong>of</strong> organized system.<br />

To overcome such issue, the Ministry <strong>of</strong> Health formed a committee for Breast<br />

Cancer Prevention and Early Detection to implement a program. In addition a<br />

National Society for Cancer Prevention was formed and it included many <strong>of</strong> this<br />

center board members. Furthermore, Cancer Prevention and Early Detection<br />

Symposia were held targeting primary health care physicians.<br />

Important barriers include screening-accompanied anxiety in waiting for further<br />

studies or confirmation <strong>of</strong> findings. To eliminate and shorten the period <strong>of</strong> time<br />

from screening until physician visit, we opened a special clinic for those referred<br />

from the center, and a coordinator assigned to open a file and facilitate the referral<br />

procedure in order to follow their referral to the hospital within a week from their<br />

referral to the tertiary center.<br />

In order to make sure that patients with suspicious lesions are properly managed,<br />

a flow system was established as illustrated in figures.<br />

Annual<br />

Follow-up<br />

Mammography<br />

Figure 1: Figure Patient 1: Patient flow diagram for for participants in the breast in the cancer breast screening cancer program. screening<br />

program.<br />

Table 1: Targeted Barriers for the Screening Program and Interventions<br />

Targeted Barriers Intervention<br />

Financial - Initial donation<br />

- Endowment<br />

Confidentiality - All women staff, closed space<br />

- ID for identification<br />

- Secure Database Site<br />

Staff shortage - 2 radiologists to read<br />

mammography for back-up<br />

Difficulty in retrieving data and data - Establishing database with new<br />

management<br />

s<strong>of</strong>tware<br />

Lack <strong>of</strong> interest/misinformation from - Public awareness program<br />

patient and physician<br />

implemented throughout the year<br />

- Physician education symposia and<br />

5<br />

activities<br />

- High-pr<strong>of</strong>ile media events<br />

Patient Care - Recall system<br />

- Work-up and management/referral<br />

process<br />

- Follow-up<br />

Conclusions<br />

In spite <strong>of</strong> having many barriers to public cancer screening, though we were able<br />

to screen more than 2500 women within 18 months period, many <strong>of</strong> these barriers<br />

were overcome by specific intervention. Good strategic planning with attention to<br />

the above challenges and following WHO cancer control program for implementing<br />

such center prior to establishing another center is advisable.<br />

References<br />

1. The World Health Organization Fight <strong>Against</strong> Cancer: Strategies that prevent,<br />

cure and care. 2007. www.who.int/cancer/en<br />

2. Agarwal G. et al. Breast Care 2008;3:21-27 DOI 10.1159/000115288<br />

3. E. Ibrahim. JCO Vol. 36, No. 155: May 2008;2009<br />

4. Baron-Epel. Mammography screening in a multi-ethnic population in Israel.<br />

Health Services Research. 42(3):1008-1019.2007<br />

5. Khadiga F. Dandash, Abdulrahman Al-Mohaineed. Knowledge, attributes and<br />

practicesurrounding breast cancer and screening in female teachers <strong>of</strong> Buraidah,<br />

46 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info<br />

Screening<br />

Center<br />

• Suspicious<br />

lesion<br />

R4/R5<br />

• R0<br />

• R3<br />

No Yes<br />

Annual<br />

Follow-up<br />

Mammography<br />

Referral<br />

Center for<br />

W/U<br />

Management<br />

by the Tertiary<br />

Center<br />

Negative<br />

for<br />

Malignancy<br />

Positive for<br />

Malignancy


Saudi <strong>Arab</strong>ia. International <strong>Journal</strong> <strong>of</strong> Health Science. Vol. 1, Jan 2007<br />

6. Faisal Azaiza, Mini Cohen. Devloping and testing an instrument for identifying<br />

culture-specific barriers to breast cancer screening in Israel. Acta Oncologica.<br />

Volume 47, Issue 8 2008<br />

7. Little D. et al. Breast cancer in Asian women. Ethnomed. June 2, 2001. pp<br />

1570-1577<br />

8. M. Lamyian, A. Hydrania, et al. Barriers to and factors facilitating breast<br />

cancer screening among Iranian Women: A qualitative study. Health <strong>Journal</strong><br />

Vol. 13 No 5:Sept. – Oct. 2007<br />

9. Layla A. Al-Alaboud, et al. The barriers <strong>of</strong> breast cancer screening program<br />

among PHHC female physicians. Middle East <strong>Journal</strong> <strong>of</strong> Family Medicine. Sept.<br />

2006; Vol. 6 Issue 5)<br />

ENDOMETRIAL CARCINOMA<br />

Faisal Al Safi, FRCSC 1 , Hany Salem 2 , Nashmia Al Mutairi 1<br />

(1) King Abdulaziz <strong>Medical</strong> City – National Guard Health Affairs, Riyadh, KSA<br />

(2) King Faisal Specialist Hospital and Research Center, Riyadh, KSA<br />

Corresponding Author: Dr. Faisal Al Safi , FRCSC, Department <strong>of</strong> <strong>Oncology</strong><br />

(Mail Code 1777), P.O. Box 22490, Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

E-mail: safif@ngha.med.sa<br />

Endometrial carcinoma is the most common gynecologic malignancy in the United<br />

States; it accounts for 6 percent <strong>of</strong> all cancers in women. Fortunately, most cases<br />

are diagnosed at an early stage when surgery alone may be adequate for cure.<br />

Five-year survival rates for localized, regional, and metastatic disease are 96, 67,<br />

and 26 respectively [1].<br />

Risk Factors<br />

Estrogen<br />

Treatment with estrogen alone increases the risk for endometrial hyperplasia and<br />

carcinoma. Endometrial hyperplasia can be demonstrated within one year in 20 to<br />

50 percent <strong>of</strong> women receiving unopposed estrogen [2]. Furthermore, any factor<br />

that increase exposure to estrogen[e.g., hormone replacement therapy, obesity,<br />

anovulatory cycles. estrogen–secreting tumor]increases the risk <strong>of</strong> endometrial<br />

cancer, whereas factor that decrease exposure to estrogens or increase progesterone<br />

level[e.g. oral contraceptives or smoking ] tend to be protective [3].<br />

Tamoxifen<br />

Tamoxifen is a competitive inhibitor <strong>of</strong> estrogen binding to estrogen receptors<br />

that also has partial agonist activity (i.e., tamoxifen is a weak estrogen). It is used<br />

for adjuvant therapy in women with early stage breast cancer, as treatment for<br />

recurrent disease, and for reduction <strong>of</strong> breast cancer incidence in high-risk women.<br />

The site-specific activity <strong>of</strong> tamoxifen in different tissues is well recognized,<br />

suppressing the growth <strong>of</strong> breast tissue, but stimulating the endometrial lining.<br />

Tamoxifen use has been linked to development <strong>of</strong> endometrial pathology, both<br />

benign and malignant [4].<br />

Diabetes and Hypertension<br />

Women with diabetes mellitus and hypertension are at increased risk for endometrial<br />

cancer, at least in part because <strong>of</strong> co-morbid factors associated with obesity.<br />

However, some studies have found independent effects as well [5].<br />

Diet<br />

A diet containing high amounts <strong>of</strong> fat (especially animal fat) appears to be a risk<br />

factor for endometrial cancer, even after adjusting for caloric intake and body<br />

weight [6]. By comparison, two case-control studies showed that plant-based diets<br />

high in fiber, legumes (especially soybeans [7]), whole grain foods, vegetables,<br />

and fruits appear to reduce the risk <strong>of</strong> the disease [8].<br />

Other risk factors include:<br />

Familial predisposition and lack <strong>of</strong> physical activity.<br />

Pathology<br />

The most common type <strong>of</strong> endometrial cancer is endometriod adenocarcinoma (75<br />

to 80 percent). Clear cell and papillary serous carcinomas account for 1 to 5, and<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 47


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

5 to 10 percent <strong>of</strong> endometrial cancer cases respectively. Mucinous and squamous<br />

cell cancer comprises less than 2 percent <strong>of</strong> endometrial cancers.<br />

Clinical Features<br />

Ninety percent <strong>of</strong> patient with endometrial carcinoma have abnormal vaginal<br />

bleeding, most commonly postmenopausal bleeding. Occasionally, vaginal<br />

bleeding does not occur because <strong>of</strong> cervical stenosis, particularly in thin, elderly,<br />

estrogen-deficient patients. In some patients with cervical stenosis , a hematometria<br />

develops, and a small percentage have a purulent vaginal discharge resulting<br />

from pyometria.<br />

Diagnosis<br />

Endometrial biopsy<br />

Endometrial biopsy is the initial diagnostic test to rule out endometrial cancer<br />

in women with abnormal uterine bleeding or endometrial cells on Pap smear.<br />

Dilatation and Curettage with hysteroscopy and directed biopsy should be<br />

considered when the endometrial biopsy is nondiagnostic, but there remains a<br />

high suspicion <strong>of</strong> cancer (e.g., hyperplasia with atypia, presence <strong>of</strong> necrosis,<br />

pyometra, or persistent bleeding).<br />

Transvaginal ultrasonography<br />

Transvaginal ultrasound can be used to evaluate the endometrium by measuring the<br />

endometrial wall thickness. In postmenopausal women, an endometrial thickness<br />

<strong>of</strong> less than 4 to 5 mm is associated with a low risk <strong>of</strong> endometrial disease [9-10];<br />

a thicker lining should be further evaluated by <strong>of</strong>fice biopsy, hysteroscopy with<br />

directed biopsy, or D&C. Cancer becomes increasingly more frequent relative to<br />

benign disease as the endometrial thickness approaches 20 mm, which was the<br />

mean endometrial thickness in 759 women with endometrial cancer [10] .<br />

Management<br />

The cornerstone <strong>of</strong> treatment for endometrial cancer is total abdominal hysterectomy<br />

and bilateral salpingo-oophorectomy, and this operation should be performed in<br />

all cases whenever feasible. In addition, many patients require some type <strong>of</strong><br />

adjuvant radiation therapy to help prevent vaginal vault recurrence and to sterilize<br />

occult disease in lymph nodes. Chemotherapy in endometrial cancer is only <strong>of</strong><br />

palliative value [11].<br />

Screening<br />

General population<br />

Screening for endometrial cancer is generally not warranted in asymptomatic<br />

women. Many cases are diagnosed at an early stage since the malignancy<br />

commonly causes abnormal vaginal bleeding. Moreover, adequate inexpensive,<br />

noninvasive screening tests are not currently available. Although endometrial cancer<br />

occasionally is diagnosed after endometrial or glandular cells are discovered on<br />

Pap smear, the sensitivity <strong>of</strong> the Pap smear for detection <strong>of</strong> endometrial cancer is<br />

low and not sufficient to recommended it as a screening tool. Endometrial biopsy<br />

is more sensitive, but is relatively uncomfortable; equivocal tests may lead to<br />

additional unnecessary evaluation [12].<br />

Women at risk <strong>of</strong> Hereditary Nonpolyposis Colorectal Cancer (HNPCC)<br />

Women who are at risk <strong>of</strong> HNPCC are also at high risk (40 to 60 percent) <strong>of</strong><br />

developing endometrial cancer. In fact, the risk <strong>of</strong> developing endometrial cancer<br />

may be slightly higher than the risk <strong>of</strong> developing colon cancer and endometrial<br />

cancer may be the first manifestation <strong>of</strong> malignancy [13]. These women also have<br />

a 12 percent lifetime risk <strong>of</strong> developing ovarian cancer [14].<br />

Because <strong>of</strong> the high risk for development <strong>of</strong> endometrial cancer in women with or<br />

at risk <strong>of</strong> HNPCC, the American Cancer Society recommends that annual screening<br />

by endometrial biopsy be initiated by age 35 [15]. These recommendations cover<br />

the following:<br />

• Women who are known to carry HNPCC-associated mutations<br />

• Women who have a family member known to carry this mutation<br />

• Women from families with an autosomal dominant predisposition to colon<br />

cancer in the absence <strong>of</strong> genetic testing.<br />

At present, there are no data regarding the efficacy <strong>of</strong> this approach, nor is there<br />

a consensus on the optimal age (25 to 35) to begin screening. Annual or biennial<br />

pelvic ultrasonography has not been shown to be effective for early detection <strong>of</strong><br />

endometrial cancer in these populations [16]<br />

These women should also be counseled about preventive measures, such as the<br />

option <strong>of</strong> prophylactic hysterectomy at complete childbirth.<br />

Patient on Tamoxifen<br />

The American College <strong>of</strong> Obstetricians and Gynecologists has developed the<br />

following recommendations for monitoring women on tamoxifen [17];1<br />

• Perform an annual gynecologic examination.<br />

• Monitor for symptoms <strong>of</strong> endometrial hyperplasia or cancer. Women<br />

should be educated to report any abnormal vaginal symptoms (e.g. , bloody<br />

discharge, spotting, staining, leukorrhea).<br />

• Investigate any abnormal vaginal symptoms.<br />

• Limit tamoxifen use to five years duration because benefit beyond this time<br />

has not been demonstrated.<br />

• If atypical endometrial hyperplasia develops, the use <strong>of</strong> tamoxifen should<br />

be reassessed and appropriate gynecologic management should be initiated.<br />

Hysterectomy should be considered for women with atypical endometrial<br />

hyperplasia in whom tamoxifen therapy must be continued.<br />

Conclusion<br />

Endometrial carcinoma is the most common gynecologic malignancy; to date<br />

there is no rule for screening program in general population. However, high risk<br />

patient for endometrial cancer have to be evaluated and followed up independently.<br />

Recommendation and Guidelines for Screening and Prevention <strong>of</strong> the<br />

Endometrial Cancer<br />

Screening<br />

The committee members (Authors) agreed that from reviewing all the data currently<br />

available regarding the role <strong>of</strong> screening for the general population <strong>of</strong> the women<br />

for endometrial cancer, it clearly emphasize that there is no role for such screening<br />

program in detecting early or pre-invasive changes for those women.<br />

Initiating such screening program using either vaginal ultrasound with or without<br />

endometrial sampling will not be cost effective, and its effect on the morbidity<br />

and mortality for those women will be extremely low and will not be beneficial<br />

for the society.<br />

Early Detection<br />

As there is no rule for screening for the general population, the committee members<br />

48 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


agreed that the best approach for such a condition will be the education for the<br />

general population and for the primary care physician and family physician<br />

regarding the early symptoms and signs <strong>of</strong> endometrial cancer.<br />

Any women who present with post menopausal or Premenopausal bleeding or<br />

abnormal uterine bleeding should be referred immediately to her gynecologist<br />

in order for her to do the necessary work-up which must include endometrial<br />

sampling at least in order to explore the development <strong>of</strong> hyperplasia or malignancy.<br />

The committee members agreed that for special sub-group which represent high<br />

risk for developing endometrial cancer, like those who have history <strong>of</strong> breast<br />

cancer and they are on Tamoxifen, or those women with chronic anovulation, they<br />

should have regular check-up by gynecologist, where they will have ultrasound<br />

to evaluate the thickness <strong>of</strong> the endometrial lining according to gynecologist<br />

assessment and their symptoms and the decision for endometrial sampling will<br />

be left for the gynecologist according to the patient ultrasound result and her<br />

symptoms. Also, these women should be aware to report to their physician when<br />

they have abnormal bleeding and endometrial samples or dilation and curratage<br />

(D & C) must be done on these patients.<br />

References<br />

1. Jemal A, Murray T, Waard W, et al. cancer statistics,2005,CA Cancer J clin<br />

2005;55:10<br />

2. Woodruff JD, Pickar JH. Incidence <strong>of</strong> Endometrial hyperplasia in postmenopausal<br />

women taking conjugated estrogens [premarin] with medroxyprogesterone acetate<br />

or estrogens alone. Am J obstetgynecol 1994;170:12-13<br />

3. Parazzini F, LaVecchia C, Bocciolone L, Franceschi S, The epidemiology <strong>of</strong><br />

endometrial cancer. Gynecol oncol 1991; 41:1-16<br />

4. Gohen I. Endometrial pathologies associated with postmenopausal tamoxifen<br />

treatment .Gynecol oncol 2004; 94:256.<br />

5. Soler M, Chatenoud L, Negri E, etal.Hypertension and Hormone-related<br />

neoplasm's in women. Hypertension 1999;34:320<br />

6. Potischman N, Swanson CA, Brinton LA, etal. Dietary association in case-control<br />

study <strong>of</strong> endometrial cancer. Cancer Causes Control1993; 4:239.<br />

7. Xu, WH, Zheng, W, Xiang, VB, etal. Soya food intake and risk <strong>of</strong> endometrial<br />

cancer among Chinese women in shanghai: population based case-control study.<br />

BMJ 2004; 328:1285.<br />

8. Goodman MT, Wilkens LR, Hankin JH, etal. <strong>Association</strong> <strong>of</strong> soy and fiber<br />

consumption with the risk <strong>of</strong> endometrial cancer. AMJ Epidemiology 1997;146:294<br />

9. Goldstein SR, Nachtigall M, Snyder JR, etal. Endometrial assessment by vaginal<br />

ultrasonography before endometrial sampling in patient with postmenopausal<br />

bleeding.Am J obstet Gynecol 1990; 163:119.<br />

10. Karlsson B, Granberg S, Wikland M, etal. Transvaginal ultrasonogrophy <strong>of</strong><br />

the endometrium in women with postmenopausal bleeding. aNordic multicenter<br />

study . Am J obstet Gynecol 1995; 172:1488.<br />

11. Neville F. Hacher. 2005. Uterine cancer. Practical gynecology oncology.<br />

411-429<br />

12. Koss LG, Schreiber K, Oberlander SG, etal. Detection <strong>of</strong> endometrial carcinoma<br />

and hyperplasia in asymptomatic women .Obstet Gynecol 1984, 64:1.<br />

13. Lu, KH, Dinh M, Kohlmann W, etal. Gynecologic cancer as a sentinel cancer<br />

for women with hereditary nonpolyposis colorectal cancer syndrome.Obstet<br />

Gynecol 2005; 105:569.<br />

14. Aarnio M, Sankila R, Pukkala E, etal. Cancer risk in mutation carriers <strong>of</strong><br />

DNA-mismatch-repair genes.Int J Cancer 1999; 81:214.<br />

15. Smith RA, Von Eschenbach, AC, Wender R, etal. American cancer society<br />

guidelines for the early detection <strong>of</strong> cancer: update <strong>of</strong> early detection guidelines<br />

for prostate, colorectal and endometrial cancer.CA Cancer J Clin 2001; 51:38.<br />

16. Dove-Edwin I, Boks D, G<strong>of</strong>f S, etal. The out come <strong>of</strong> endometrial carcinoma<br />

surveillance by ultrasound scan in women at risk <strong>of</strong> hereditary nonpolyposis<br />

colorectal carcinoma and familial colorectal carcinoma. Cancer 2002;94:170<br />

17. American College <strong>of</strong> Obstetricans and Gynecologists. Tamoxifen and<br />

endometrial cancer .AGOG Committee opinion, AGOG2000.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 49


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

OVARIAN CANCER<br />

Faisal Al Safi, FRCSC 1 , Haney Salem 1 , Nashmia Al Mutairi 1<br />

(1) King Abdulaziz <strong>Medical</strong> City – National Guard Health Affairs, Riyadh, KSA<br />

Corresponding Author: Dr. Faisal Al Safi , FRCSC<br />

Department <strong>of</strong> <strong>Oncology</strong> (Mail Code 1777)<br />

P.O. Box 22490 Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

E-mail: safif@ngha.med.sa<br />

Introduction<br />

Ovarian cancer remains a highly lethal disease. Although it is the second most<br />

common female reproductive cancer, preceded by cancer <strong>of</strong> the uterine corpus,<br />

more women die from ovarian cancer than from cervical and uterine cancer<br />

combined. The principle reason for these poor outcomes is the advance stage <strong>of</strong><br />

disease at diagnosis in 70-75% <strong>of</strong> cases and an overall 5-year survival <strong>of</strong> only<br />

20-30%. However, women with a diagnosis <strong>of</strong> stage 1 disease achieve a 90-95%<br />

probability <strong>of</strong> cure (1)<br />

Risk factors<br />

The risk <strong>of</strong> epithelial ovarian cancer increase with age, especially around the<br />

time <strong>of</strong> menopause. A family history <strong>of</strong> epithelial ovarian cancer is one <strong>of</strong> the<br />

most important risk factor, infertility and not bearing children are also risk factor,<br />

while pregnancy and use <strong>of</strong> birth control pills can decrease the risk <strong>of</strong> developing<br />

epithelial ovarian cancer. The risk factors for stromal cell and germ cell tumor<br />

are unknown (2).<br />

Pathology<br />

The majority (90%) <strong>of</strong> primary ovarian cancer derived from epithelial cells.<br />

Although they can also arise from other cell type (germ cell tumor,sex cord stromal<br />

tumor ,and mixed cell type tumor).(2)<br />

Epithelial ovarian cancer [EOC] constitutes different histological subtype, <strong>of</strong> which<br />

serous type is the most prevalent 60% <strong>of</strong> all EOC. Other type include mucinous,<br />

endometroid, clear cell, brener, and mixed phenotype tumor, in addition to their<br />

distant morphological appearance and subtle clinical differences, there is molecular<br />

evidence for heterogeneity between different EOC subtype [ 3].<br />

Clinical symptoms and diagnosis<br />

The best way to detect early ovarian cancer is to have a high index <strong>of</strong> suspicion <strong>of</strong><br />

the diagnosis in the symptomatic women, persistent symptoms such as an increase<br />

in abdominal size, abdominal pain and bloating, fatigue, indigestion, inability<br />

to eat normally, urinary frequency, pelvic pain, constipation, back pain, urinary<br />

incontinence <strong>of</strong> recent oncent, or unexplained weight loss should be evaluated with<br />

ovarian cancer being included in differential diagnosis. Because ovarian cancer<br />

occurs most frequently in the postmenopausal women (median age approximately<br />

60 years), these symptoms should not be ignored. Unfortunately, many clinician<br />

and patient quick to attribute such symptoms to menopause, aging, dietary changes,<br />

stress, or functional bowel problems. As a result, delays <strong>of</strong> weeks or months<br />

<strong>of</strong>ten occur before medical advice is sought or diagnostic studies are performed.<br />

In evaluating this symptom physical examination including a pelvic examination<br />

and imaging studies including vaginal ultrasonography may be helpful in making<br />

the diagnosis. CA125 measurement has not been shown to be useful in most<br />

circumstances because elevated levels <strong>of</strong> CA125 are associated with a variety<br />

<strong>of</strong> common benign condition .in post menopausal women with a pelvic mass,<br />

a CA125 measurement may be helpful in predicting a higher likelihood <strong>of</strong> a<br />

malignant tumor than a benign tumor, a normal CA125 measurement alone dose<br />

not rule out ovarian cancer because up to 50% <strong>of</strong> early stage cancers and 20-25%<br />

<strong>of</strong> advance cancers are associated with normal values (1,4 ).<br />

Treatment<br />

In contrast to other types <strong>of</strong> cancer, surgery is always considered for women<br />

with both localized and advanced ovarian cancer. Surgery is necessary for both<br />

accurate staging and optimal cytoreduction , and is crucial in successful treatment<br />

<strong>of</strong> this disease. The combination <strong>of</strong> optimal cytoreductive surgery and effective<br />

chemotherapy has led to significant improvements in survival for women with<br />

ovarian cancer. (2)<br />

Screening<br />

To date, no screening techniques; including CA125 level measurement and pelvic<br />

ultrasonography, have been proven effective in screening low risk asymptomatic<br />

women for ovarian cancer.(4)<br />

For high risk patients, annual gynecologic examination with annual pelvic<br />

examination is recommended. Hereditary ovarian cancer is estimated to be<br />

represent only 5-10% <strong>of</strong> all ovarian cancers. Based on current data, a woman with<br />

a germ line mutation <strong>of</strong> BRCA1 or BRCA2 has a life risk <strong>of</strong> 15-45% <strong>of</strong> developing<br />

ovarian cancer. There is no data demonstrating that screening improves early<br />

detection <strong>of</strong> ovarian cancer in this population. These women should be <strong>of</strong>fered<br />

genetic counseling to address issues that relate to their high risk <strong>of</strong> breast and<br />

ovarian cancer and the potential impact <strong>of</strong> these genetic mutations on their <strong>of</strong>fspring.<br />

Even if this group were screened for ovarian cancer on regular basis, more than<br />

90% <strong>of</strong> all potential ovarian cancer patients would remain unscreened. (1)<br />

Prophylactic salpingo-oopherctomy has been shown to decrease the risk <strong>of</strong> breast<br />

as and gynecological cancer in women with inherited risk for these malignancies.<br />

However, multiple questions remain unanswered regarding this procedure. (5)<br />

Conclusion<br />

Ovarian cancer is the most common cause <strong>of</strong> death in women from gynecological<br />

malignancy, there is no role <strong>of</strong> screening test in general population, high risk<br />

patient need annual gynecologic examination and counseling.<br />

Recommendations and Guidelines for Screening and Prevention <strong>of</strong> Ovarian Cancer<br />

• The committee members ( Authors) ; after reviewing all the current available<br />

data from most <strong>of</strong> the studies conducted on the role <strong>of</strong> screening for a general<br />

population for ovarian cancer, concluded that there is no place for initiating<br />

a screening program for the women in the general population as it is proven<br />

from all the studies conducted on this issue that it will not be cost effective,<br />

and also its effect on the morbidity and mortality from the disease is very<br />

limited, as a large number <strong>of</strong> patient will be unnecessarily screened and<br />

operated on in order to detect one early case <strong>of</strong> ovarian cancer.<br />

• For that reason, the committee members agreed that the best approach is to<br />

try to identify patients who are at high risk for ovarian cancer. Those women<br />

should be identified as women with high risk for familial ovarian cancer,<br />

50 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


and counseling should be done for them, and they should be included in a<br />

screening program designed specifically for the high risk group which will<br />

include the following:<br />

1. Annual pelvic examination by a gynecologist;<br />

2. Annual pelvic ultrasound with colored Doppler for the ovaries; and<br />

3. Annual CA 125 levels<br />

• For the very high risk patients who have more than two first degree related<br />

or one first degree related and two second degree relatives, it is advised<br />

for those patients to do the BRCA 1 and BRCA 2 study, to counsel those<br />

patients and advise them regarding the advantage <strong>of</strong> performing prophylactic<br />

oophorectomy.<br />

• The committee members feel that it is very important to educate all<br />

gynecologists regarding the issue <strong>of</strong> familial ovarian cancer where it is<br />

very important to identify the patients at high risk, where they should advise<br />

the patient regarding enrolment in this screening program, and advise them<br />

regarding this issue, and their first and second degree relatives for the risk <strong>of</strong><br />

familial malignancies, and encouraging involvement in a screening program.<br />

References<br />

1.AGOG committee opinion no280, December2002.<br />

2. Jonathan S. Berek, Neville F. Haker. Epithelial ovarian cancer.practical<br />

Gynecologic oncology 2004; 4:443-494.<br />

3. Scully RE. Pathology <strong>of</strong> ovarian cancer precursors. J cell Biol 1995,<br />

23(suppl):208-18.<br />

4. Jacobs I, Davier AP, Bridges J, etal. Prevalance screening for ovarian cancer<br />

in post menopsaulal women by CA125 measurement and ultrasonography.BMJ<br />

1993; 306:1030-1034.<br />

5. SGO Committee statement on prophylactic salpingo-oophorectomy. Gynecologic<br />

oncology 98(2005)179-181.<br />

CERVICAL CANCER<br />

Faisal Al Safi, FRCSC 1 , Haney Salem 1 , Nashmia Al Mutairi 1<br />

(1) King Abdulaziz <strong>Medical</strong> City – National Guard Health Affairs, Riyadh, KSA<br />

Corresponding Author: Dr. Faisal Al Safi , FRCSC<br />

Department <strong>of</strong> <strong>Oncology</strong> (Mail Code 1777)<br />

P.O. Box 22490, Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

E-mail: safif@ngha.med.sa<br />

Introduction<br />

Cervical cancer is the second most common cause <strong>of</strong> cancer-related morbidity<br />

and mortality among women in developing countries. It Usually affects women<br />

between ages <strong>of</strong> 30 and 55 but has been found as early as the teen years and<br />

uncommonly after age <strong>of</strong> 75(1).<br />

Risk factor<br />

The major risk factors for cervical cancer include early onset <strong>of</strong> sexual activity,<br />

multiple sexual partners, and a high-risk sexual partner (e.g., promiscuous sexual<br />

activity, sexual exposure to a partner with human papillomavirus infection<br />

[2]). Other risk factors are a history <strong>of</strong> sexually transmitted diseases (eg,<br />

Chlamydia trachomatis, herpes simplex virus) [3], smoking [4], high parity [5],<br />

immunosuppression, low socioeconomic status, and previous history <strong>of</strong> vulvar<br />

or vaginal squamous dysplasia.<br />

Pathology<br />

Squamous cell carcinomas (SCCs) account for approximately 80 percent <strong>of</strong><br />

cervical cancers, adenocarcinomas 15 percent, and adenosquamous carcinomas<br />

3 to 5 percent. Small cell carcinomas and other types are rare (6)<br />

Clinical manifestations<br />

Early cervical cancer is frequently asymptomatic, the most common symptoms<br />

at presentation are:<br />

• Abnormal vaginal bleeding<br />

• Post coital bleeding<br />

• Vaginal discharge that may be watery, mucoid, or purulent and malodorous<br />

Pelvic or lower back pain, which may radiate along the posterior side <strong>of</strong> the lower<br />

extremities, can occur with advanced disease. Bowel or urinary symptoms, such<br />

as pressure-related complaints, hematuria, hematochezia, or vaginal passage <strong>of</strong><br />

urine or stool, are uncommon and suggest advanced disease. (7)<br />

Diagnosis<br />

Diagnosis <strong>of</strong> cancer is confirmed by biopsy in women with a grossly visible lesion.<br />

Symptomatic women without a visible lesion and those who have only abnormal<br />

cervical cytology should undergo colposcopy with directed biopsy or, if necessary,<br />

diagnostic conization. In addition, any cervix that is unusually firm or expanded<br />

should be sampled by punch biopsy and endocervical curettage, even if the cervical<br />

cytology smear does not show evidence <strong>of</strong> neoplasia. Histologic confirmation <strong>of</strong><br />

invasive cervical cancer is followed by a careful staging evaluation that should<br />

include a thorough physical examination. The cervix and entire vagina should be<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 51


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

carefully inspected and palpated to identify overt tumor or subepithelial vaginal<br />

extension. Rectovaginal examination permits the best assessment <strong>of</strong> tumor size and<br />

parametrial involvement. Palpation <strong>of</strong> the liver and inguinal and supraclavicular<br />

lymph nodes is important to screen for metastatic disease (7).<br />

Treatment<br />

Treatment <strong>of</strong> invasive cervical cancer involves management <strong>of</strong> both the primary<br />

leision and potential site <strong>of</strong> metastatic disease. Both surgery or and chemo<br />

radiotherapy may be used for treatment according to clinical stage and risk <strong>of</strong><br />

recurrence.<br />

HPV Vaccine<br />

Human Papillomavirus Vaccine (HPV) is a vaccine that prevents infection with<br />

certain species <strong>of</strong> Human Papillomavirus associated with the development <strong>of</strong><br />

cervical cancer and genital wart.<br />

Two HPV Vaccines are currently available:<br />

Gardasil and Cervarix. Both vaccine protect against two <strong>of</strong> the HPV types (16,<br />

18) that cause cervical cancer, and some other genital cancer, Gardasil also protect<br />

against two <strong>of</strong> the HPV types (6, 11) that cause genital wart (8).<br />

Although available data for both vaccines is promising. Still long term efficacy<br />

and safety is unknown.<br />

Screening<br />

Cervical cytology screening programs can detect preinvasive, as well as invasive,<br />

cellular changes <strong>of</strong> the cervix. Because cervical cancer typically has a long<br />

preinvasive state (<strong>of</strong>ten a decade or more) and the treatment for preinvasive<br />

disease is effective, screening programs potentially can prevent the occurrence<br />

<strong>of</strong> invasive cervical cancer. [9]. although cervical cancer accounts for relatively<br />

few deaths in the United States, it is one <strong>of</strong> the leading causes <strong>of</strong> cancer death in<br />

women in developing countries. This observation is thought to be directly related<br />

to the lack <strong>of</strong> screening programs in those areas.<br />

A variety <strong>of</strong> screening guidelines have been proposed; the choice depends upon<br />

available resources (11, 12, 13).<br />

Recommendation and Guidelines for Screening and Prevention <strong>of</strong> Cervical Ca<br />

All the committee members (Authors) strongly recommend the initiation <strong>of</strong> a<br />

structured, well-organized screening program for cervical cancer among <strong>Arab</strong><br />

women.<br />

• The design for the program should be suitable and acceptable for our<br />

community and for the cultural aspects <strong>of</strong> our women in the <strong>Arab</strong> World.<br />

• It is recommended that the screening process should be done by the Pap smear<br />

using liquid base media in agreement with the international recommendation.<br />

• The screening process should start one year after the women get married and<br />

start her sexual activity and the screening should go on in accordance with the<br />

international recommendation which is once annually, then if the patient has<br />

three consecutive normal Pap smear, she can do Pap smear every three years.<br />

• For the high risk population, the screening should continue on annual basis<br />

in order to prevent any development <strong>of</strong> invasive disease.<br />

• Regarding the addition <strong>of</strong> HPV test to the screening process, the committee<br />

members feel that a cost effectiveness study should be conducted first in<br />

order to evaluate the prevalence <strong>of</strong> the HPV between the Saudi women<br />

at different age groups, also to determine the sub-groups which are more<br />

prevalent within our society.<br />

According to the results <strong>of</strong> such study, the decision will be made if the<br />

addition <strong>of</strong> HPV testing after the age <strong>of</strong> 30 for those women will be cost<br />

effective and will be beneficial for them or not.<br />

• Regarding the issue <strong>of</strong> vaccination for the Saudi girls between the ages <strong>of</strong><br />

9 to 26, the committee members agreed that this issue should be dealt with<br />

individually at the current time, as there are many social and economic<br />

issues connected to it.<br />

The decision regarding the vaccination should be left to the family regarding<br />

their choices for the immunization <strong>of</strong> their daughters after getting all the<br />

necessary information from their physician.<br />

The committee members agreed that without the cohort study on the prevalence <strong>of</strong><br />

PHV in Saudi women, the recommendations for vaccinating girls before marriage<br />

will lack cost effectiveness and benefits before we can get the results <strong>of</strong> such study.<br />

References<br />

1. Parkin DM, Pisani P, Ferlay J. Global cancer statistics .CA cancer J1999; 49:33.<br />

2. Castellsage X, Bosch X, Munoz N, etal. Male circumcision, penile human<br />

papillomavirus infection, and cervical cancer in female partners. N Engl J Med<br />

2002; 346:1105.<br />

3. Hawes SE, Kiviat NB, Are genital infections and inflammation c<strong>of</strong>actors in<br />

the pathogenesis <strong>of</strong> invasive cervical cancer?.J Natl cancer inst 2002; 94:1592.<br />

4. Catle PE, Wacholder S, Lorincz AT. etal. A prospective study <strong>of</strong> high grade<br />

cervical neoplasia risk among human papilloma virous-infected women. J Natl<br />

cancer inst 2002; 94:1406.<br />

5. Munoz N, Franceschi S, etal. Role <strong>of</strong> parity and HPV in cervical cancer: the<br />

IARC multicenter case-control study.Lancet 2002; 359:1093.<br />

6. Alboes-saavedra J, Gersell D, etal. Terminology <strong>of</strong> endocrine tumors <strong>of</strong><br />

the uterine cervix: results <strong>of</strong> a worshop sponsored by the collage <strong>of</strong> Amirican<br />

pathologists and the National cancer insutute.Arch pathol lab med 1997; 121:34.<br />

7. Jonathan S. Berek, Neville F. Haker. Cervical cancer. Practical Gynecologic<br />

oncology.2004; 4:337-372.<br />

8. Cummins J. Recombinant Cervical Cancer Vaccines, Science in Society 29;<br />

20-21, 2006<br />

9. Womack Warren AY. Achievable laboratory standards: a review <strong>of</strong> cytology <strong>of</strong><br />

99 women with cervical cancer. Cytopathology 1998; 9:171.<br />

10. Saslow D, etal. American Cancer Society Guidelines for the Early Detection<br />

<strong>of</strong> Cervical Neoplasia and Cancer. CA Cancer J Clin 2002; 52:342-362<br />

11. USPSTF. Screening for Cervical Cancer. Jan 2003.<br />

12. ACOG. Cervical Cytology Screening. ACOG Practice Bulletin no. 45. ACOG<br />

2003;102: 417-427<br />

52 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


ESOPHAGEAL CANCER<br />

Abdul-Rahman Jazieh, MD, MPH, Omalkhair Abulkhair, MD<br />

King Abdulaziz <strong>Medical</strong> City for National Guard, Riyadh, KSA<br />

Corresponding Author: Abdul Rahman Jazieh, MD, MPH<br />

Department <strong>of</strong> <strong>Oncology</strong> (Mail code 1777)<br />

P.O. Box 22490, Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

E-mail: jazieha@ngha.med.sa<br />

Introduction<br />

Esophageal cancer is a common cancer which has poor prognosis with a five year<br />

survival <strong>of</strong> less than 15%. Although squamous cell cancer is the most common<br />

histologic subtype, adenocarcinoma is increasing rapidly.<br />

This manuscript will present the epidemiology and risk factors <strong>of</strong> esophageal<br />

cancer and review early detection and prevention <strong>of</strong> related issues.<br />

Epidemiology<br />

Esophageal cancer ranks 6th among cancers worldwide (constitutes 5.7% <strong>of</strong><br />

all cancer mortality). It does increase with age with peak between age 50 – 70.<br />

(Globocan 2002). Esophageal cancer is more common in male; with male to<br />

female ratio <strong>of</strong> 3-5:1.<br />

Risk Factors<br />

Smoking and Alcohol<br />

Smoking and alcohol are major risk factors for esophageal cancer (UTD 14 – 15).<br />

Smoking does not only increase the risk <strong>of</strong> squamous cell carcinoma but also<br />

increase the risk <strong>of</strong> adenocarcinoma especially in patients with Barretts esophagus<br />

as the risk is about 2.4 times higher that non smokers (UTD 13).<br />

Gastroesophageal Reflux and Barrett’s Esophagus<br />

Gastroesophageal reflux is a major risk factor for adenocarcinoma as the risk <strong>of</strong><br />

esophageal cancer increases significantly in patients with GE reflux. The odd ratio<br />

<strong>of</strong> having esophageal cancer is more than 7 times for patients with reflux symptoms<br />

which may increase to more than 40 times if the symptom is long standing for<br />

more than 20 years (79-80).<br />

Underlying Esophageal Diseases<br />

Such achalasia and caustic esophageal injury increases the risk <strong>of</strong> esophageal<br />

cancer (43, 44).<br />

Obesity and Dietary Risk Factors<br />

Eating or drinking hot food or drinks may cause increase in the risk <strong>of</strong> esophageal<br />

cancer e.g. hot tea (29), food that contains U-nitroso may increase the risk <strong>of</strong><br />

esophageal cancer so is the consumption <strong>of</strong> Betel nuts.<br />

Furthermore, low selinum and zinc diets have been implicated in increasing the<br />

risk <strong>of</strong> esophageal cancer. Obesity with BMI between 25 – 30 kg/m2 increase the<br />

odd ratio <strong>of</strong> esophageal cancer by 1.5 and if the BMI was more than 30 kg/m2,<br />

the odd ratio increased to 2.7 (83,84).<br />

Medication Use<br />

The use <strong>of</strong> oral biphosphenate and medications that decrease the lower esophageal<br />

sphincter (e.g. nitroglycerine, aminophyllin) increase the risk <strong>of</strong> esophageal cancer.<br />

Presenting Sign and Symptoms<br />

The symptoms <strong>of</strong> esophageal cancer result from the obstructive lesion that causes<br />

dysphagea especially <strong>of</strong> the solid food, odanophagea, regungitation, etc. These<br />

may lead into anorexia and weight loss.<br />

Patient may develop respiratory symptom due to aspiration or tracheoesophageal<br />

fistula. Chroninc blood loss or frank bleeding will lead to anemia and its associated<br />

symptoms.<br />

Screening<br />

There is no study demonstrated the value <strong>of</strong> mass screening for esophageal cancer.<br />

Management <strong>of</strong> Esophageal Cancer<br />

Early esophageal cancer is managed by surgical resection and combined<br />

multidiscipline treatment with radiation therapy and/or chemotherapy.<br />

Advanced stages are usually non curable and the goal <strong>of</strong> care will be to alleviate<br />

the obstructive symptoms.<br />

Prevention<br />

Avoidance <strong>of</strong> alcohol and smoking is paramount to decrease the risk <strong>of</strong> esophageal<br />

cancer. Treating esophageal reflux would help to increase fiber intake may have<br />

protective value.<br />

Esophageal Cancer Prevention<br />

Primary Prevention<br />

• Avoid smoking and alcohol consumption.<br />

• Treat esophageal reflux.<br />

Public Screening<br />

• No evidence <strong>of</strong> benefit for public screening.<br />

Reference<br />

1. Globocan 2002: http://www.dep.iarc.fr<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 53


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

LUNG CANCER PREVENTION AND EARLY DETECTION<br />

Abdul-Rahman Jazieh, MD, MPH<br />

King Abdulaziz <strong>Medical</strong> City for National Guard, Riyadh, KSA<br />

Corresponding Author: Abdul Rahman Jazieh, MD, MPH<br />

Department <strong>of</strong> <strong>Oncology</strong> (Mail code 1777)<br />

P.O. Box 22490, Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

E-mail: jazieha@ngha.med.sa<br />

Abstract<br />

Lung cancer is the most common cancer in incidence and mortality worldwide.<br />

Most lung cancer cases are attributed to tobacco use which makes it very amendable<br />

to preventative interventions.<br />

Although there is no proven benefit <strong>of</strong> mass screening for lung cancer, large study<br />

about the role <strong>of</strong> spinal CT scan is ongoing. Smoking cessations and avoidance <strong>of</strong><br />

other known risk factors will reduce the risk <strong>of</strong> lung cancer significantly.<br />

This manuscript discusses the epidemiology <strong>of</strong> lung cancer, its risk factors and<br />

the value <strong>of</strong> early detection and prevention.<br />

Introduction<br />

Lung cancer ranks first in the world in incidence and mortality. Multiple risk<br />

factors have been identified and the majority <strong>of</strong> lung cancer cases are preventable.<br />

This manuscript presents summary <strong>of</strong> the epidemiology <strong>of</strong> lung cancer and risk<br />

factors and reviews its prevention and early detection recommendations.<br />

Epidemiology<br />

Lung cancer is the most common cancer worldwide (1.35 million <strong>of</strong> 10.9 million<br />

<strong>of</strong> new cases) and the deadliest cancer (1.18 million <strong>of</strong> 6.7 million cancer-related<br />

deaths).(1)<br />

As per the Saudi National Cancer Registry 2004 statistics, there were 296 cases <strong>of</strong><br />

lung cancer accounting for 4.2% <strong>of</strong> all diagnosed cases.(2) Lung cancer ranked<br />

fourth among male population and sixteenth among female population. It affected<br />

233 (78.7%) males and 63 (21.3%) females with male to female ratio <strong>of</strong> 3.7:1. The<br />

overall Adjusted Standard Rate (ASR) was 3.1/100,000. ASR was 5.5/100,000<br />

for males and 1.5/100,000 for females, which is much less than the Western<br />

figures. For example, ASR in the United States is 85.7/100,000 for males and<br />

54.2/100,000 for females. The mean age at diagnosis was 64 years among males<br />

(range 24-98 years) and 61 years among females (range 24-89 years). The most<br />

common morphological subtypes are squamous cell carcinoma, adenocarcinoma<br />

and small cell lung cancer Stage distribution showed that 55.7% are having distant<br />

metastasis at presentation, and localized disease, regional and unknown represent<br />

13.2%, 10.8% and 20.3%, respectively.<br />

Pathology<br />

Lung cancer is divided into non-small cell lung cancer (more than 80% <strong>of</strong><br />

cases) and small cell lung cancer. The non-small cell lung cancer is divided into<br />

adenocarcinoma, squamous cell and large cell carcinoma.<br />

There was a shift in the incidence <strong>of</strong> squamous cell carcinoma and adenocarcinoma.<br />

Up to the late 1980, squamous cell lung cancer was the most common subtype,<br />

which was then surpassed by adenocarcinoma.<br />

It is note worthy that the risk <strong>of</strong> all <strong>of</strong> these subtype <strong>of</strong> cancer including<br />

adenocarcinomas is increased by smoking. This is contrary to the thought <strong>of</strong> some<br />

practitioners that adenocarcinoma risk does not increase by smoking which is driven<br />

by the fact the adenocarcinoma is the most common sub-types in non-smokers.<br />

Risk Factors for Lung Cancer<br />

a. Smoking<br />

The rapid increase <strong>of</strong> lung cancer over the last century from a rare disease<br />

to an epidemic is attributed to the exposure to newly introduced major risk<br />

factors, which include smoking at the top <strong>of</strong> the list. Around 85-90% <strong>of</strong> lung<br />

cancer cases could be attributed to the use <strong>of</strong> tobacco, directly or indirectly. (3)<br />

The relative risk <strong>of</strong> developing lung cancer is 11 – 20 times more in smokers<br />

compared to non-smoker. The risk <strong>of</strong> lung cancer is dependent on the number<br />

<strong>of</strong> cigarette smoked per day (calculated by pack/year number) and the duration<br />

<strong>of</strong> smoking with increase in risk <strong>of</strong> smoking started at younger age. (4 - 6)<br />

For example, 35 year old man has a 9% chance <strong>of</strong> dying from lung cancer<br />

before age 85 if he smokes less than 25 cigarettes per day. This risk increases<br />

to 18% if he smokes more than 25 cigarette per day. (7)<br />

The environmental tobacco exposure (ETS), which may be referred to as<br />

“second hand smoking”, increases the risk by 27-80%. (8 - 11) ETS may occur<br />

at home or at work. For example, the risk <strong>of</strong> non-smoker spouse increases by<br />

20 – 30% if the spouse is smoker over non-smoker’s spouse. (12,13) About<br />

25% <strong>of</strong> the lung cancer in non-smoker is attributed to second hand smoking<br />

which constitutes about 5% <strong>of</strong> all lung cancer cases.<br />

b. Radon Gas<br />

The exposure to radon is an established risk factor <strong>of</strong> lung cancer, which<br />

was initially observed in uranium miners. (14, 15) However, this naturally<br />

occurring radioactive gas is a delay product <strong>of</strong> uranium – 238 and radium –<br />

226 and accumulates also in buildings and homes especially in basements<br />

and lower level floors.<br />

The exposure to the indoor radon may be responsible for up to 9% if lung<br />

cancer in Western countries and it has synergistic effect with smoking.<br />

Smoking in miners increases the risk <strong>of</strong> lung cancer by 10 times over the<br />

non-smoker miners.(16)<br />

c. Industrial and Occupational Exposure<br />

Exposure to various carcinogens has been linked to lung cancer. The list <strong>of</strong><br />

these carcinogens includes: arsenic, polycyclic hydrocarbons, diesel exhaust,<br />

herbicides and insecticides, silica, asbestos, beryllium and chromium.<br />

Asbestos is well known cause <strong>of</strong> not only mesothelioma but also <strong>of</strong> primary<br />

lung cancer. The risk <strong>of</strong> exposure to asbestos is about 5 times more than<br />

the general population but when it is combined with smoking, a synergetic<br />

effect takes place and increases the risk up to 50 – 100 times. The incidence<br />

peak <strong>of</strong> cancer occurs 25 – 30 years after exposure. (17 - 21)<br />

d. Air Pollution<br />

Exposure to outdoor pollution, especially nitrogen oxides from the traffic<br />

fumes has been linked to increase risk <strong>of</strong> lung cancer. (10, 22)<br />

54 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


e. Other Risk Factors<br />

There are other risk factors that were associated with increase incidence<br />

<strong>of</strong> lung cancer including family history, sedentary life, alcohol and dietary<br />

factors with variable strength <strong>of</strong> association. (23 - 26)<br />

Presenting Signs and Symptoms<br />

Patients with lung cancer present with three categories <strong>of</strong> manifestations like most<br />

solid tumors (27 - 28) The first type is related to the mass effect <strong>of</strong> the primary<br />

tumors including: cough, chest pain, shortness <strong>of</strong> breath and hemophysis, postobstructive<br />

pneumonia and superior vena cava syndrome.<br />

The second type is related to distant metastatic lesions which may include<br />

seizure, pathologic fracture, lymphadenopathy or organomegaly. The third type<br />

<strong>of</strong> manifestations is related to systemic paraneoplastic manifestations not related<br />

to the mass effect per se which include: hypercalcemia, hyponateremia, cushing<br />

syndrome, neurological manifestation, weight loss, or digital clubbing (hypertrophic<br />

pulmonary osteoarthropathy).<br />

Management <strong>of</strong> Lung Cancer<br />

The management <strong>of</strong> lung cancer should be based on multidisciplinary team<br />

approach. The treatment is usually stage dependent. For early stages I, II and<br />

selected III: surgery is the main approach. Adjuvant chemotherapy is helpful in<br />

resected stages II and III but not stage I.<br />

For stage III A/B which is considered locally advanced disease, treatment is<br />

usually combined chemotherapy and radiotherapy with surgical intervention in<br />

selected cases. For metastatic disease: systemic therapy using chemotherapy or<br />

biological therapy is the standard approach.<br />

Prognosis and Outcome<br />

The prognosis <strong>of</strong> lung cancer is stage dependent with higher survival rates for<br />

earlier stages. The 5 years survival for stages ranges from 67% for stage I to less<br />

than 1% in stage IV. (32)<br />

Therefore, the earlier the cancer is discovered, the better the chance <strong>of</strong> survival.<br />

Hence, the importance <strong>of</strong> identifying an effective screening methods would be<br />

<strong>of</strong> great value.<br />

Screening and Early Detection<br />

Various studies using chest x-ray, sputum cytology and spinal CT scans were not<br />

supportive <strong>of</strong> routine mass screening. (29 - 31) A large multisite National Cancer<br />

Institute-USA sponsored study <strong>of</strong> spinal CT scan including more than 50,000<br />

participants may help answer this question.<br />

Prevention<br />

Smoking cessation at any age is <strong>of</strong> proven benefits <strong>of</strong> reduction <strong>of</strong> lung cancer<br />

risk over extended period <strong>of</strong> time (15 – 20 years) but it remains higher than never<br />

smoker risk. (8, 34 - 35)<br />

If smoker cannot quit completely, reducing the number <strong>of</strong> cigarette smoked may<br />

reduce the risk <strong>of</strong> cancer. (36) Smoking cessation and eliminating the risk <strong>of</strong><br />

tobacco will eradicate the majority <strong>of</strong> lung cancer cases making it one <strong>of</strong> the most<br />

preventable cancer. (8, 35, 37) It is imperative to have a systemic campaigns or<br />

tobacco control programs that ban public advertising and promotions, especially<br />

those which targets the youth and that ban smoking in public areas such as<br />

restaurants or workplace. (38 - 44) Minimizing the occupational exposure to<br />

the above mentioned carcinogens will decrease the risk for lung cancer further.<br />

Conclusion<br />

While lung cancer is the leading cancer in incidence and mortality, it is also a<br />

preventable disease in the majority <strong>of</strong> cases.<br />

Recommendations for Early Detection and Prevention for Lung Cancer<br />

Primary Prevention:<br />

• Cancer prevention is highly recommended by avoiding tobacco products<br />

(including cigarettes, cigars, chewing tobacco, arkila or shisha and passive<br />

indirect smoking) smoking cessations and avoiding environmental tobacco<br />

exposure and exposure to other known risk factors.<br />

• Stopping all forms <strong>of</strong> tobacco promotion and advertising is a major component<br />

<strong>of</strong> lung cancer prevention.<br />

Early Detection:<br />

• No method was proven efficacious in early detection <strong>of</strong> lung cancer that<br />

translated into better survival.<br />

References<br />

1. Parkin D, Bray F, et al. Global cancer statistics 2002. Ca Cancer J 2005;55:74-<br />

108.<br />

2. National Cancer Registry, 2004. Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia.<br />

3. Shopland D. Tobacco use and its contribution to early cancer mortality with<br />

a special emphasis on cigarette smoking. Environ Health prospect 1995:103;<br />

131-142.<br />

4. Doll R, Peto R, Boreham J, et al. Mortality from cancer in relation to smoking:<br />

50 years observations on British doctors. Br J Cancer 2005; 92:426-9.<br />

5. Doll R, Peto R. Cigarette smoking and brochial carcinoma: dose and time<br />

relationships among regular smokers and lifelong non-smokers. J Epidemiol<br />

Community Health 1978; 32:303 – 13.<br />

6. Wiencke JK, Thurston SW, Kelsey KT, et al. Early age at smoking initiation and<br />

tobacco carcinogen DNA damage in the lung. J Natl Cancer Inst 1999;91:614-9.<br />

7. Mattson ME, Pollack ES, Cullen JW. What are the odds that smoking will kill<br />

you? Am J Public Health 1987; 77:425-31.<br />

8. Crispo A, Brennan P, Jockel KH, et al. The cumulative risk <strong>of</strong> lung cancer among<br />

current, ex- and never-smokers in European men. Br J Cancer 2004;91:1280-6.<br />

9. Miller DP, De Vivo I, Neuberg D, et al. <strong>Association</strong> between sel-reported<br />

environmental tobacco smoke exposure and lung cancer: modification by GSTP1<br />

polymorphism. Int J Cancer 2003; 104: 758-63.<br />

10. Vineis P, Airoldi L, Veglia F, et al. Environmental tobacco smoke and risk <strong>of</strong><br />

respiratory cancer and chronic obstructive pulmonary disease in former smokers<br />

and never smokers in the EPIC prospective study. BMJ 2005; 330:227.<br />

11. Wen W, Shu XO, Gao YT, et al. Environmental tobacco smoke and mortality<br />

in Chinese women who have never smoked: prospective cohort study. BMJ<br />

2006;333:376.<br />

12. Fontham ET, Correa P, Reynolds P, et al. Environmental tobacco smoke and<br />

lung cancer in nonsmoking women. A multicenter study. JAMA 1994; 271:1752-9.<br />

13. Alberg A, Samet J. Epidemiology <strong>of</strong> lung cancer. Chest 2003;123:21S-49S.<br />

14. IARC. Ionizing radiation, part 2: some internally deposited radionuclides. Views<br />

and expert opinions <strong>of</strong> an IARC working group on the evaluation <strong>of</strong> carcinogenic<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 55


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

risks to human. Lyon, 14 – 21 June 2000. IARC Monogr Eval Carcinog Risks<br />

Hum 2001; 78:1-559.<br />

15. Frumkin H, Samet JM. Radon. CA Cancer J Clin 2000:51:337-44.<br />

16. Darby S, Hill D, Auvinen A, et al. Radon in homes and risk <strong>of</strong> lung cancer:<br />

collaborative analysis <strong>of</strong> individual data from 13 European case-control studies.<br />

BMJ 2005;330:223.<br />

17. Hammond EC, Selik<strong>of</strong>f IJ, Seidman H. Asbestos exposure, cigarette smoking<br />

and death rates. Ann N Y Acad Sci 1979; 330:473-90.<br />

18. Berry G, Lidell FD. The interaction <strong>of</strong> asbestos and smoking in lung cancer:<br />

a modified measure <strong>of</strong> effect. Ann Occup Hyg 2004; 48:459-62.<br />

19. Chen TM, Kuschner WG. Non-tobacco related lung carcinogens. Lung cancer<br />

principle and practice. Harvey pass et al. Lippincot Williams and Wilkins, Third<br />

edition, 2005:61-73.<br />

20. Lee PN. Relation between exposure to asbestos and smoking jointly and the<br />

risk <strong>of</strong> lung cancer. Occup Environ Med 2001;58:145-53.<br />

21. Lidell FD. The interaction <strong>of</strong> asbestos and smoking in lung cancer. Ann Occup<br />

Hyg 2001; 45:341-56.<br />

22. Nafstad P, Haheim LL, Wisl<strong>of</strong>f T, et al. Urban air pollution and mortality in a<br />

cohort Norwegian men. Environ Health Perspect 2004; 112:610-5.<br />

23. Liu Y, Sobue T, Otani T, et al. Vegetables, fruit consumption and risk <strong>of</strong> lung<br />

cancer among middle-aged Japanese men and women: JPHC study. Cancer<br />

Causes Control 2004; 15:349-57.<br />

24. Freudenheim JL, Ritz J, Smith-Warner SA, et al. Alcohol consumption and risk <strong>of</strong><br />

lung cancer: a pooled analysis <strong>of</strong> cohort studies. Am J Clin Nutr 2005; 82:657- 67.<br />

25. Tardon A, Lee WJ, Delgado-Rodriguez M, et al. Leisure-time physical activity<br />

and lung cancer: a meta-analysis. Cancer Causes Contro 2005; 16:389-97.<br />

26. Nitadori J, Inoue M, Iwasaki M, et al. <strong>Association</strong> between lung cancer<br />

incidence and family history <strong>of</strong> lung cancer: data from a large-scale populationbased<br />

cohort study, the JPHC study. Chest 2006; 130: 968-75.<br />

27. Spiro SG, Gould MK, Colice GL. Initial evaluation <strong>of</strong> the patient with lung<br />

cancer: symptoms, signs, laboratory, tests and paraneoplastic syndromes.<br />

ACCP Evidenced-Base Clinical Practice Guidelines (2nd Edition) Chest 2007;<br />

132:149S-160S<br />

28. Scagliotti G. Symptoms, signs and staging <strong>of</strong> lung cancer. Eur Respir Mon<br />

2001; 17:86-119<br />

29. Henschke CI, McCauley DI, Yahkelvitz DF, et al. Early Lung Cancer Action<br />

Project: overall design and findings from baseline screening. Lancet 1999;<br />

354:99-105.<br />

30. Swensen SJ, Jett JR, Sloan JA, et al. Screening for lung cancer with low-dose<br />

spiral computed tomography. Am J Resp Crit Care Med 2002; 165:508-513.<br />

31. Bach FH, Kelley M, Tate RC, et al. Screening for lung cancer: a review <strong>of</strong> the<br />

literature. Chest 2003; 123:72S-82S.<br />

32. Mountain CF. Revisions in the International System for Staging lung cancer.<br />

Chest 1997; 111:1710-7.<br />

33. Newcomb PA, Carbone PP. The Health consequences <strong>of</strong> smoking: cancer. Med<br />

Clin North Am 1992; 76:305-31.<br />

34. Halpern MT, Gillespie BW, Warner KE. Patterns <strong>of</strong> absolute risk <strong>of</strong> lung cancer<br />

mortality in former smokers. J Natl Cancer Inst 1993;85:457-64.<br />

35. Peto R, Darby S, Deo H, et al. Smoking, smoking cessation, and lung cancer<br />

in UK since 1950: combination <strong>of</strong> national statistics with two case-control studies.<br />

BMJ 2000;321: 323-9.<br />

36. Godtfredsen NS, Prescott E, Osler M. Effect <strong>of</strong> smoking reduction on lung<br />

cancer risk. JAMA 2005; 294:1505-10.<br />

37. Ebbert JO, Yang P, Vachon CM, et al. Lung cancer risk reduction after smoking<br />

cessation: observations from a prospective cohort <strong>of</strong> women. J Clin Oncol 2003;<br />

21:921-926.<br />

38. Homan CD, Donovan RJ, Corti B, et al. Banning tobacco sponsorship: replacing<br />

tobacco with health messages and creating health-promoting environments. Tob<br />

Control 1997;6:115-21.<br />

39. Farelly MC, Evans WN, Sfekas AE. The impact <strong>of</strong> workplace smoking bans:<br />

results from a national survey. Tob Control 1999;8:272-7.<br />

40. McVey D, Stapleton J. Can anti-smoking television advertising affect smoking<br />

behaviour? Controlled trial <strong>of</strong> the Health Education Authority for England’s antismoking<br />

TV campaign. Tob Control 2000;9:273-82.<br />

41. Wakefield M, Chaloupka F. Effectiveness <strong>of</strong> comprehensive tobacco control<br />

programmes in reducing teenage smoking in the USA. Tob Control 2000;9:177-86.<br />

42. Braverman MT. Adolescent smoking and exposure to tobacco marketing under<br />

a tobacco advertising ban: findings from 2 Norwegian national samples. Am J<br />

Public Health 2004;94:1230-8.<br />

43. Shields M. Smoking bans: influence on smoking prevalence. Health Rep<br />

2007;18:9-24.<br />

44. Bala M. Strzeszynski L, Cahill K. Mass media interventions for smoking<br />

cessation in adults. Cochrane Database Syst Rev 2008; 23:(1): CD004704.<br />

56 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


ORAL MALIGNANCIES:<br />

ROLE OF PREVENTION AND EARLY DETECTION<br />

Dr. Abdulaziz Binahmed<br />

King Abdulaziz <strong>Medical</strong> City, Riyadh<br />

Corresponding Author: Dr. Abdulaziz Binahmed, BDS, MDent, MSc, FRCDC<br />

Consultant, Department <strong>of</strong> Surgery, King Abdulaziz <strong>Medical</strong> City<br />

P.O. Box 22490 Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

Assistant Pr<strong>of</strong>essor, Department <strong>of</strong> Surgery<br />

University <strong>of</strong> Manitoba, Winnipeg, Canada<br />

E-mail: azizbinahmed@mac.com<br />

Introduction<br />

The oral cavity is that part <strong>of</strong> the upper aerodigestive tract which extends from the<br />

mucocutaneous junction at the vermillion border <strong>of</strong> the lip to the anterior surface<br />

<strong>of</strong> the faucial arch. It is lined by squamous epithelium containing interspersed<br />

minor salivary glands and it also contains dento-alveolar structures that support<br />

the upper and lower dentition. Primary tumors <strong>of</strong> the oral cavity may arise from<br />

the surface epithelium, minor salivary glands or submucous s<strong>of</strong>t tissue as well as<br />

from dental structure, bone or neurovascular tissue. Squamous cell carcinoma <strong>of</strong><br />

the oral cavity forms more than 90% <strong>of</strong> all newly diagnosed cases <strong>of</strong> oral cancer,<br />

with the majority <strong>of</strong> patients being males. A rising incidence has been observed in<br />

females over the past 50 years. Anatomical sites in the oral cavity that have been<br />

described by the International Union <strong>Against</strong> Cancer (UICC)/ American Joint<br />

Committee on Cancer (AJCC) Staging System are the tongue, floor <strong>of</strong> mouth,<br />

gingiva, buccal mucosa, retromolar trigone and hard palate. The tongue and the<br />

floor <strong>of</strong> mouth are the most common sites <strong>of</strong> origin for primary squamous cell<br />

carcinoma in the oral cavity in the Western World. In other parts <strong>of</strong> the world<br />

e.g. Middle East and Asia, the retromolar trigone and buccal mucosa are the most<br />

frequently encountered primary sites due to tobacco chewing and the chewing<br />

<strong>of</strong> betel nuts. Despite controversy and debate during the last century between<br />

different health care disciplines regarding the best treatment <strong>of</strong> oral cancer using<br />

radiation, surgery or chemotherapy, surgery still plays a primary role in the control<br />

<strong>of</strong> the disease.<br />

Disease Epidemiology<br />

Oral cavity cancers account for about 3% <strong>of</strong> all cancers diagnosed each year in<br />

north America. (1,2) This is estimated to be 27,000 newly diagnosed cases in the<br />

United States and 3,200 cases in Canada. Slightly more than 10,000 Americans<br />

and 1,000 Canadians will die <strong>of</strong> oral cancer each year. Studies from around the<br />

globe show that for both sexes combined cancer <strong>of</strong> the mouth and pharynx ranks<br />

sixth overall behind lung, stomach, breast, colon, rectum and cervix uteri in that<br />

order. The rates range from a low <strong>of</strong> 1.8/100,000 per year to a high <strong>of</strong> 47/100,000<br />

per year. The highest rates <strong>of</strong> oral cancer in the world are found in France, the<br />

Indian subcontinent, Brazil and central/eastern Europe. There are also marked<br />

differences between countries in the same geographic regions.<br />

The incidence <strong>of</strong> oral cancer increases with age in all parts <strong>of</strong> the world. In the West,<br />

98% <strong>of</strong> the patients are over 40 years <strong>of</strong> age. In the high prevalence areas <strong>of</strong> the world,<br />

many <strong>of</strong> the patients are less than 35-years-old owing to heavy usage <strong>of</strong> various<br />

forms <strong>of</strong> tobacco. Furthermore, it is now clear that in many Western countries,<br />

there has been an alarming rise in the incidence <strong>of</strong> oral cancer during the past two or<br />

three decades particularly among younger men a trend that appears to be continuing.<br />

In industrialized countries, men are affected two to three times as <strong>of</strong>ten as women.<br />

The most important risk factors are alcohol and tobacco consumption for intraoral<br />

cancer and sun exposure for lip cancer in those who work outdoors. The incidence<br />

<strong>of</strong> tongue and other intraoral cancer for woman can be greater or equal to that <strong>of</strong><br />

men in high incidence areas such as India where chewing tobacco is also common<br />

among women. There has been a gradual increase <strong>of</strong> the number <strong>of</strong> female patients<br />

reflected by the change in male to female ratio in the Western societies.<br />

The reported head and neck cancer cases in Saudi <strong>Arab</strong>ia by the National Cancer<br />

Registry for the year 2002 was approximately 700 new cases, (3) and we believe<br />

that this number is increasing annually as the aging population is increasing.<br />

Risk Factors<br />

Tobacco<br />

There is absolutely no doubt that on a global scale the use and abuse <strong>of</strong> tobacco<br />

products is the major cause <strong>of</strong> oral cancer. Typically 90% <strong>of</strong> men and 60% <strong>of</strong><br />

women with oral carcinomas use tobacco. The incidence rate <strong>of</strong> oral carcinoma<br />

in smokers is six to ten times greater when compared to non-smokers.<br />

Alcohol<br />

It is very difficult to separate the effects <strong>of</strong> alcohol and tobacco as most heavy<br />

alcohol consumers also use tobacco. Nevertheless, some cohort and case control<br />

studies have found an increased risk <strong>of</strong> upper aerodigestive tract cancer associated<br />

with alcohol drinking in non-smokers. The epidemiological evidence shows that<br />

all <strong>of</strong> alcoholic drinks are dangerous if heavily consumed.<br />

Viruses<br />

The knowledge <strong>of</strong> viruses as a human carcinogen has improved in the past two<br />

to three decades. Viruses contribute to the multi-step process <strong>of</strong> carcinogenesis<br />

in many human neoplasms. Human papilloma virus, especially human papilloma<br />

16, is the most common type associated with both cervical and oral cancer. In<br />

vitro studies show that high-risk HBV types can immortalize primary human<br />

oral epithelial cells.<br />

Presenting Signs and Symptoms<br />

The common presenting symptoms include pain, oral ulcer, oral mass, and neck<br />

mass. While the common presenting signs include leukoplakia, erythroplakia,<br />

exophytic mass, oral ulcer and neurological alterations.<br />

Outcome & Prognosis<br />

Despite the improvements in surgical and radiotherapeutic techniques, intraoral<br />

squamous cell carcinoma has relatively unfavorable prognosis with an overall<br />

five-year survival rate <strong>of</strong> 35 – 50%. The survival rate has regrettably remained<br />

virtually unchanged over the past three decades. The increase in the incidence<br />

<strong>of</strong> oral cancer accentuates this mortality from the cancer.<br />

Several parameters have been adapted and applied by clinicians to evaluate the<br />

prognosis <strong>of</strong> oral cancer. These parameters can be divided into epidemiological<br />

parameters which include age, sex, race, alcohol and tobacco intake and<br />

comorbidity; clinical parameters which include the TNM classification, stage,<br />

and the site <strong>of</strong> the primary tumor; and histological parameters which include the<br />

marginal status, the perineural or perivascular invasion, histopathological grading,<br />

tumor thickness and extracapsular spread.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 57


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

The single most important factor affecting long-term results after treatment <strong>of</strong><br />

carcinoma <strong>of</strong> the oral cavity is the stage <strong>of</strong> the disease at the time <strong>of</strong> presentation.<br />

For early stage tumors excellent cure rate is achieved. The five year survival rate<br />

for patients with oral cancer treated at Memorial Sloan-Kettering Cancer Center<br />

between 1986 and 1995 showed that for Stage I oral cancer the five year diseasespecific<br />

survival was more than 90% while for Stage II, III, IV 80%, 65%, 55%,<br />

respectively. The overall survival in the same center was site dependent; with<br />

tumors in the buccal mucosa and retromolar trigone having the worst outcome. (4)<br />

Magnano et al(5,6) found that the T stage was a consistent and dependent predictor<br />

<strong>of</strong> pathologically involved cervical lymph nodes. In addition, maximal tumor<br />

diameter has been shown to predict local recurrence in tumors arising from the<br />

lower lip, oral cavity and oropharynx. Pernot in 1996 reported on 565 patients in<br />

which he showed that the five-year survival for patients with T1 lesion was 70%<br />

compared to 29% in patients with T3 disease(7).<br />

It is not surprising that the presence <strong>of</strong> clinically positive lymph nodes at the time<br />

<strong>of</strong> presentation is probably the single most important factor in determining the<br />

outcome and prognosis. In general patients presenting with neck node metastases<br />

do half as well as patients who present with a primary tumor only. Jones et al(8)<br />

performed a univariate and multivariate analysis <strong>of</strong> a number <strong>of</strong> tumor factors<br />

and prognosis in oral cancer on 524 patients. Their study showed that only the T<br />

stage and the N stage were significant as predictors for survival.<br />

Mamelle et al(9) reviewed 914 patients and his multivariate analysis showed that the<br />

number <strong>of</strong> positive nodes was a significant and independent predictor <strong>of</strong> survival.<br />

Olsen et al(10) has shown that the number <strong>of</strong> positive lymph nodes reflects the<br />

prognosis and overall survival in 284 patients that he studied. This information is<br />

in agreement with a study that Shah(11) performed on 704 patients showing that<br />

the failure rate was significantly higher as the number <strong>of</strong> positive nodes increased.<br />

Screening Methods Overview<br />

A routine Head and neck examination in any patient above the age <strong>of</strong> forty who has<br />

any <strong>of</strong> the risk factors mentioned earlier. A focused oral examination is mandatory<br />

with proper inspection and palpation <strong>of</strong> all the high-risk areas in high risk patients.<br />

Prevention<br />

Primary prevention is the approach that concentrates on eliminating the risk factors.<br />

Educating the public on these risk factors is the first step in primary prevention.<br />

Although educating the primary health care providers on approaches to eliminate<br />

or at least reduce risk factors such as smoking and alcohol consumption.<br />

Secondary prevention is the approach that concentrates on early detection <strong>of</strong> the<br />

disease (Screening). In oral cancer, this approach is preformed via routine clinical<br />

and oral examination for high risk patients (Smokers, >40 years, Alcohol, Poor<br />

Diet) as general public screening in oral cancer is not cost effective.<br />

Tertiary prevention is the approach that focus on reducing recurrence <strong>of</strong> the treated<br />

disease or minimize treatment morbidity.<br />

Recommendations for Early Detection and Prevention for Head and Neck<br />

Primary Prevention:<br />

• Cancer prevention is highly recommended by avoiding tobacco products<br />

by smoking or chewing.<br />

• Avoiding alcoholic drinks.<br />

Early Detection:<br />

• No method was proven efficacious in early detection <strong>of</strong> oral cavity cancers.<br />

• Good clinical exams for high-risk patients; smokers older than 40 years,<br />

alcohol consumption and poor nutrition.<br />

References<br />

1. Jemal A, Murray T, Samuels A, et al: Cancer statistics, 1999. CA Cancer J<br />

Clin 53:5,2003.<br />

2. Canadian cancer statistics 2004.<br />

3. Cancer Incidence Report, Saudi <strong>Arab</strong>ia, 2002. National Cancer Institute.<br />

4. Shah JP. Head and neck surgery, 2nd ed. London: Mosby-Wolfe, 1996: 189-196<br />

5. Magnano M, De Stefani A, Lerda W, Usai A, Ragona R, Bussi M, Cortesina G.<br />

Prognostic factors <strong>of</strong> cervical lymph node metastasis in head and neck squamous<br />

cell carcinoma. Tumori. 1997 Nov-Dec;83(6):922-6.<br />

6. Magnano M, Bongioannini G, Lerda W, Canale G, Tondolo E, Bona M, Viora<br />

L, Gabini A, Gabriele P. Lymphnode metastasis in head and neck squamous cells<br />

carcinoma: multivariate analysis <strong>of</strong> prognostic variables. J Exp Clin Cancer Res.<br />

1999 Mar;18(1):79-83<br />

7. Pernot M, H<strong>of</strong>fstetter S, Peiffert D, Aletti P, Lapeyre M, Marchal C, Luporsi<br />

E, Bey P, Nancy VL. Role <strong>of</strong> interstitial brachytherapy in oral and oropharyngeal<br />

carcinoma: reflection <strong>of</strong> a series <strong>of</strong> 1344 patients treated at the time <strong>of</strong> initial<br />

presentation. Otolaryngol Head Neck Surg. 1996 Dec;115(6):519-26.<br />

8. Jones AS. Prognosis in mouth cancer: tumor factors Eur J Cancer B Oral<br />

Oncol. 1994 Jan;30B(1):8-15.<br />

9. Mamelle G, Pampurik J, Luboinski B, Lancar R, Lusinchi A, Bosq J. Lymph<br />

node prognostic factors in head and neck squamous cell carcinomas in patients<br />

with lymph node involvement. Am J Surg. 1994 Nov;168(5):494-8.<br />

10. Olsen KD, Caruso M, Foote RL, Stanley RJ, Lewis JE, Buskirk SJ, Frassica<br />

DA, DeSanto LW, O'Fallon WM, Hoverman VR Primary head and neck cancer.<br />

Histopathologic predictors <strong>of</strong> recurrence after neck dissection. . Arch Otolaryngol<br />

Head Neck Surg. 1994 Dec;120(12):1370-4.<br />

11. Shah JP, Cendon RA, Farr HW, Strong EW. Carcinoma <strong>of</strong> the oral cavity.<br />

factors affecting treatment failure at the primary site and neck. Am J Surg. 1976<br />

Oct;132(4):504-7.<br />

58 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


URINARY BLADDER CANCER<br />

Dr. Alaa Kandil 1<br />

(1) Alexandria School <strong>of</strong> Medicine, Alexandria, Egypt<br />

Corresponding Author: Dr. Alaa Kandil<br />

Pr<strong>of</strong>essor <strong>of</strong> Clinical <strong>Oncology</strong>, Alexandria School <strong>of</strong> Medicine<br />

Alexandria, Egypt<br />

E-mail ad: alaakandil@hotmail.com<br />

Bladder cancer is the second most common genitourinary malignancy. The<br />

American Cancer Society estimated that 61,420 new cases <strong>of</strong> bladder cancer<br />

would be diagnosed in the United States during 2006 and about 13,060 individuals<br />

would die <strong>of</strong> the disease.(1)<br />

Many patients with bladder cancer experience recurrence but do not die <strong>of</strong> the<br />

disease. While bladder cancer is only the fourth most common cancer in men after<br />

lung, colorectal and prostate cancers, in terms <strong>of</strong> incidence it is the second most<br />

prevalent malignancy in middle-aged and elderly men after prostate cancer.(2),(3)<br />

The current standard <strong>of</strong> care for detecting and monitoring bladder tumors is<br />

cystoscopy, voided urine cytology and imaging.(4)<br />

However, cystoscopy is invasive, painful and costly. Therefore, it is not suitable as a<br />

screening test. Although urine cytology is a noninvasive test, it is limited by its low<br />

sensitivity <strong>of</strong> 20% to 40% for low grade tumors. Several methods have been reported<br />

for the early detection <strong>of</strong> bladder cancer using various potential markers. (5– 8)<br />

The specificity and sensitivity <strong>of</strong> these tests vary between 50% and 100%. Therefore,<br />

they are not adequate for screening patients. Ideally a urine based bladder tumor<br />

marker would be noninvasive, inexpensive and nonuser dependent, and have<br />

high accuracy. Optimal markers would serve for screening, initial diagnosis, and<br />

monitoring recurrence and progression as well as predicting prognosis.<br />

The criteria defined by Wilson and Junger for assessing the performance <strong>of</strong> a<br />

screening program appear suitable to use when discussing the role <strong>of</strong> a urinarybased<br />

assay for bladder cancer (BCa) [9].<br />

These criteria can be divided into those related to the population involved, the<br />

disease being studied, the test used, and the health economic consequences <strong>of</strong><br />

screening.<br />

It must first be stated that population screening for BCa, even with a noninvasive<br />

urinary-based test, is unlikely to be cost effective or to produce significant reductions<br />

in mortality. At least two well-described population screening programs have been<br />

reported and in both the overall incidence <strong>of</strong> BCa was 1.2–1.3% [10, 11-13].<br />

Of these few cases, around 45% were high grade and half <strong>of</strong> these already had<br />

muscle invasion. Thus, the capacity to improve outcomes using screening in the<br />

general population is low, given the few patients with early invasive tumours.<br />

However, populations at high risk <strong>of</strong> BCa can be defined because many etiologic<br />

risk factors are known. For example, cigarette smoking increases the risk <strong>of</strong> BCa<br />

4-fold and occupational exposure to chemical carcinogens may account for 20%<br />

(or more) <strong>of</strong> all tumours [14].<br />

Targeting high-risk populations with BCa screening could produce significant<br />

reductions in mortality at a frequency to justify the expense. The majority <strong>of</strong> use for<br />

a urinary assay for BCa would be in surveillance for patients previously diagnosed<br />

with the disease. These patients are compliant (because they are anxious about<br />

their disease state) and would welcome the replacement <strong>of</strong> invasive cystoscopic<br />

examination.<br />

For a screening or surveillance test to be beneficial it must identify the disease<br />

at a stage where treatment significantly improves the prognosis. A urinary-based<br />

assay that can diagnose BCa whilst confined to the urothelium or carcinoma in<br />

situ could fulfil this criterion. However, the majority <strong>of</strong> bladder tumors belong to<br />

the noninvasive phenotype.<br />

For these tumours early diagnosis, before the onset <strong>of</strong> symptoms, is unlikely to<br />

alter overall survival rates, even if it does reduce recurrence rates and morbidity (by<br />

treating lower-volume cancers). It is mainly tumors <strong>of</strong> the invasive pathway (around<br />

25–33% <strong>of</strong> all BCa) that would benefit from early diagnosis and treatment. In these<br />

cancers a clear relationship exists between stage at diagnosis and outcome[15,16]<br />

in a patient presenting with symptoms. Although evidence suggests this relationship<br />

persists in a screened [10] population, studies suggest it may not hold true in<br />

surveyed patients [17].<br />

Table 1: Current evaluated urinary assays for bladder cancer<br />

Category Target Function Sensitivity Specificity<br />

Soluble - Haemoglobin/ Oxygen carriage 50-100% Poor<br />

urinary red<br />

Proteins blood cells<br />

- Complement<br />

Immune system 50-100% 64-100%<br />

factor H- Nuclear structure 50-100% 75-90%<br />

related protein<br />

- NMP-22 Nuclear structure<br />

- BLCA-1 protein<br />

- BLCA-4 Nuclear structure 96% 100%<br />

- Survivin protein<br />

- Cytokeratins Antiapoptotic 100% 87-100%<br />

8,18,19,20 protein<br />

- Hyaluronic Cytoskeletal 82-87% 55-70%<br />

acid and - structure<br />

hyaluronidase Glycosaminoglycan 88-94% 84%<br />

Cancer<br />

cell-based<br />

assays<br />

- Cytology<br />

- microsatellite<br />

analysis<br />

- Telomerase<br />

- DNA<br />

methylation<br />

- FISH to<br />

chromsome<br />

3,7,17,9p21<br />

- DD 23<br />

- Karyometry<br />

- Malignant cells<br />

- Alterations in<br />

DNA microsatellite<br />

regions<br />

- Telomerase<br />

elongating enzyme<br />

- Gene regulation<br />

- Chromosomal<br />

instability<br />

- Cancer associated<br />

antibody<br />

- Chromosomal<br />

instability<br />

11-76%<br />

72-97%<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 59<br />

70-95%<br />

68-87%<br />

73-100%<br />

>90%<br />

>95%<br />

60-70%<br />

>90%<br />

33-68%<br />

Several authors have modeled the affect <strong>of</strong> introducing a urinary biomarker for<br />

BCa analysis and surveillance. Most agree that cost savings <strong>of</strong> around 20% could<br />

be made by the reduction in cystoscopic surveillance frequency or the avoidance<br />

<strong>of</strong> diagnostic cystoscopy [18,19,20]. These projections use markers with 60–70%<br />

sensitivities and higher specificities. More savings could be made by better<br />

performing markers. In Egypt, carcinoma <strong>of</strong> the bladder is the most prevalent


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

cancer, accounting for as many as 31% <strong>of</strong> all cancer cases [21]. Currently, it ranks<br />

first in males representing 16.2% <strong>of</strong> male cancer [22]. The estimated incidence in<br />

males in rural areas in Egypt is about 32 per 100.000 [23].<br />

The exact etiology <strong>of</strong> bladder cancer is still unknown. Several risk factors have<br />

been accused as being involved in its pathogenesis such as cigarette smoking [24],<br />

synthetic nitrogen fertilizers [25], organophosphate-based pesticides [26], aromatic<br />

amines [8], pelvic irradiation, cyclophosphamide, chronic cystitis, schistosomiasis<br />

[24], human papilloma virus [27], genetic predisposition, and some occupations<br />

[24]. The relative importance <strong>of</strong> such risk factors in the pathogenesis <strong>of</strong> the disease<br />

differs in different populations.<br />

The consensus <strong>of</strong> opinion and the amount <strong>of</strong> scientific evidence available from<br />

the literature don’t suggest that routine or regular screening for bladder cancer<br />

is recommended.<br />

Further work should be done for the identification <strong>of</strong> simple reliable tests that can<br />

detect the presence <strong>of</strong> the disease in high risk patients.<br />

Intervention Population Recommendation<br />

Primary Prevention public Avoid known<br />

carcinogens<br />

Early Detection public NOT recommended<br />

Early Detection - high risk population - cytology every<br />

- history <strong>of</strong> bladder 2 years<br />

cancer<br />

- cytology every 6<br />

months<br />

References<br />

1. Clinical Trials. American Cancer Society. Available at http://www.cancer.org.<br />

Accessed April 12, 2006.<br />

2. Feldamn AR, Kessler L, Myers MH and Naughton MD: The prevalence <strong>of</strong> cancer:<br />

estimates based on the Connecticut Tumor Registry. N Engl J Med 1986; 315: 1394.<br />

3. Messing EM: Urothelial tumors <strong>of</strong> the urinary tract. In: Campbell’s Urology,<br />

8th ed. Edited by PC Walsh, AB Retik, ED Vaughan Jr, AJ Wein, LR Kavoussi, AC<br />

Novick et al. Philadelphia: WB Saunders 2002; vol 4, pp 2732–2784.<br />

4. Badalament RA, Hermansen DK, Kimmel M, Gay H, Herr HW, Fair WR et<br />

al: The sensitivity <strong>of</strong> bladder wash flow cytometry, bladder wash cytology, and<br />

voided cytology in the detection <strong>of</strong> bladder cancer. Cancer Res 1987; 60: 1423.<br />

5. Burchardt M, Burchardt T, Shabsigh A, de la Taille A, Benson MC and Sawczuk<br />

I: Current concepts in biomarker technology for bladder cancers. Clin Chem<br />

2000; 46:595.<br />

6. Celis JE, Ostergaard M, Basse B, Celis A, Lauridsen JB, Ratz GP et al: Loss<br />

<strong>of</strong> adipocyte-type fatty acid binding protein and other protein biomarkers is<br />

associated with progression <strong>of</strong> human bladder transitional cell carcinomas<br />

Cancer Res 1996; 56:4782.<br />

7. Quek ML, Sanderson K, Daneshmand S and Stein JP: New molecular markers for<br />

bladder cancer detection. Curr Opin Urol 2004; 14: 259.<br />

8. Vlahou A, Schellhammer PF, Mendrinos S, Patel K, Kondylis FI, Gong L et al:<br />

Development <strong>of</strong> a novel proteomic approach for the detection <strong>of</strong> transitional cell<br />

carcinoma <strong>of</strong> the bladder in urine. Am J Pathol 2001; 158: 1491.<br />

9. Wilson JMG, Junger G. Principles and practice <strong>of</strong> screening for disease.<br />

Geneva, Switzerland: World Health Organization; 1968.<br />

10. Messing EM, Young TB, Hunt VB, et al. Comparison <strong>of</strong> bladder cancer outcome<br />

in men undergoing hematuria home screening versus those with standard clinical<br />

presentations.Urology 1995;45:387–96, discussion 96–7.<br />

11. Britton JP, Dowell AC, Whelan P. Dipstick haematuria and bladder cancer in<br />

men over 60: results <strong>of</strong> a community study. BMJ 1989;299:1010–2.<br />

12. Mayfield MP, Whelan P. Bladder tumours detected on screening: results at 7<br />

years. Br J Urol 1998;82:825–8.<br />

13. Messing EM, Young TB, Hunt VB, et al. Home screening for hematuria: results<br />

<strong>of</strong> a multiclinic study. J Urol 1992;148:289–92.<br />

14. Wallace DM. Occupational urothelial cancer. Br J Urol 1988;61:175–82.<br />

15. Stein JP, Lieskovsky G, Cote R, et al. Radical cystectomy in the treatment<br />

<strong>of</strong> invasive bladder cancer: long-term results in 1,054 patients. J Clin Oncol<br />

2001;19:666–75.<br />

16. Madersbacher S, Hochreiter W, Burkhard F, et al. Radical cystectomy for<br />

bladder cancer today—a homogeneous series without neoadjuvant therapy. J<br />

Clin Oncol 2003;21:690–6.<br />

17. Schrier BP,HollanderMP, van Rhijn BWG, Kiemeney LALM, Witjes JA.<br />

Prognosis <strong>of</strong> muscle-invasive bladder cancer: difference between primary and<br />

progressive tumours and implications for therapy. Eur Urol 2004;45:292–6.<br />

18. Lachaine J, Valiquette L, Crott R. Economic evaluation <strong>of</strong> NMP22 in the<br />

management <strong>of</strong> bladder cancer. Can J Urol 2000;7:974–80.<br />

19. Zippe C, <strong>Pan</strong>drangi L, Agarwal A. NMP22 is a sensitive cost-effective test in<br />

patients at risk for bladder cancer. J Urol 1999;161:62–5.<br />

20. Lotan Y, Roehrborn CG. Cost-effectiveness <strong>of</strong> a modified care protocol<br />

substituting bladder tumor markers for cystoscopy for the followup <strong>of</strong> patients<br />

with transitional cell carcinoma <strong>of</strong> the bladder: a decision analytical approach.<br />

J Urol 2002;167:75–9.<br />

21. Public health impact <strong>of</strong> schistosomiasis: disease and mortality. WHO Expert<br />

Committee on the Control <strong>of</strong> Schistosomiasis. Bull World Health Organ.71(6):657-<br />

662, 1993.<br />

22. Khaled HM: Systemic management <strong>of</strong> bladder cancer in Egypt: revisited; aswell<br />

as: Expert Opin Investig Drugs. J Egypt Natl Canc Inst., 17(3):127-31, 2005.<br />

23. Amal SI and El-Sebai I: Epidemiology <strong>of</strong> bladder cancer and ligand binding.<br />

In: Cancer bladder, Inc Florida Press, 1:28-32, 1983.<br />

24. American cancer society. Detailed guide: bladder cancer 2006. Available from<br />

http://www.cancer.org/docroot/CRI/content/CRI_2_4_2X_What are the risk factors<br />

for bladder cancer 44.asp<br />

25. Mensing TT, Speijers GJ, and Meulenbelt J: Health implications <strong>of</strong> exposure<br />

to environmental nitrogenous compounds. Toxicol Rev, 22 (1): 41-51, 2003.<br />

26. Webster LR, McKenzie GH and Moriarty HT: Organophosphate based<br />

pesticides and genetic damage implicated in bladder cancer. Cancer Genet.<br />

Cytogenet, 133(2): 112-117, 2002.<br />

27. LaRue H, Simoneau M and Fradet Y: Human papilloma virus in transitional<br />

cell carcinoma <strong>of</strong> the urinary bladder. Clin Cancer Res, 1:435-438, 1995.<br />

60 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


GASTRIC CANCER<br />

Omalkhair Abulkhair, MD 1 , Abdul Rahman Jazieh, MD, MPH 1<br />

(1) King Abdulaziz <strong>Medical</strong> City for National Guard, Riyadh, KSA<br />

Corresponding Author: Omalkhair Abulkhair, MD<br />

Department <strong>of</strong> <strong>Oncology</strong> (Mail code 1777)<br />

King Abdulaziz <strong>Medical</strong> City for National Guard<br />

P.O. Box 22490, Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

E-mail: abulkhairo@ngha.med.sa<br />

Epidemiology<br />

Although declined in the United States and the Western World, gastric cancer is<br />

still high in several countries around the world. Incidence exhibits significant<br />

geographic variability. The disease is most common in East Asia. High rates have<br />

been reported in Central and South America, Eastern Europe and parts <strong>of</strong> Middle<br />

East. In Japan, gastric cancer remains the most common type <strong>of</strong> cancer among men.<br />

The overall incidence <strong>of</strong> this condition has decreased in the past few decades.<br />

Nonetheless, gastric cancer remains a major public health issue as the fourth most<br />

common cancer and the second leading cause <strong>of</strong> death worldwide. In the first<br />

quarter <strong>of</strong> the 20th century, two thirds <strong>of</strong> gastric cancers were located in the antrum<br />

and the prepyloric area and only 10% arose in the caria or the esophagogastric<br />

junction. Since the 1970’s, however, adenocarcinoma <strong>of</strong> the proximal stomach<br />

has become increasingly common.1<br />

The age-adjusted incidence rate in the United States for the years 2000 to 2003<br />

was 8.1 per 100,000. Incidence among men is twice as high as among women.2<br />

Mortality rates for gastric cancer have been declining worldwide in recent decades,<br />

most prominently in the United States.3-4 The death rate from gastric cancer<br />

for black males was 2.3-fold higher than for whites for the years 1997 to 2001.5<br />

The annual number <strong>of</strong> new cases seems to be steady in recent years; in 2008, it is<br />

estimated 21, 500 Americans will be diagnosed with gastric cancer and 10, 880<br />

will die <strong>of</strong> it.6 Worldwide, gastric cancer is the fourth most common cancer.7<br />

Most cancers in the United States are advanced at diagnosis, which is reflected<br />

in an overall 5-year survival <strong>of</strong> 2.3% from 1996 to 2002.2<br />

On the contrary, in Saudi <strong>Arab</strong>ia, gastric cancer accounts for 3% <strong>of</strong> all newlydiagnosed<br />

cancer on the year 200, it ranked 10th among male and 14th among<br />

female with male to female ratio <strong>of</strong> 2:1. The overall ASR was 3.1/100,000. The<br />

median age is 65 years and stage is quite advanced regional in around 50% <strong>of</strong> area.8<br />

Highest incidence in Japan, South America and Eastern Europe adjusted worldwide<br />

is 15.62 per 100,000. Adjusted rate in Latin America is variable in frequent<br />

before age 40, twice as frequent leading cause <strong>of</strong> death from cancer worldwide<br />

in men than women.<br />

In Costa Rica, the incidence rate for men is 51.5 and 28.7 for women. In USA,<br />

the incidence has been decreasing and unexplained by the cancer has migrated<br />

proximally. Gastro-esophageal lesions are more frequent than antral lesions.<br />

Risk Factors 9-14<br />

Risk factors can be summarized as follows:<br />

1. Chronic Athropy<br />

Chronic atrophic gastritis is thought to be the initial step in the development <strong>of</strong> most<br />

gastric cancer, chronic atrophic gastritis has been shown to appear in patients with:<br />

• Tobacco use<br />

• H. Pylori infection<br />

• Diets with high level <strong>of</strong> nitrites, nitrates, salt and smoked food<br />

• Previous gastric surgery<br />

• Pernicious anemia<br />

• Alcohol<br />

Smoking increases the risk <strong>of</strong> gastric cancer by 50% to 60%. It has been estimated<br />

that smoking tobacco is responsible for 11% <strong>of</strong> all stomach cancers worldwide.<br />

Tobacco use decreases the level <strong>of</strong> caroteroids and vitamin C which act as protective<br />

agents against this disease. In addition, tobacco use associated with H. pylori<br />

infection which in turn leads to atrophic gastritis. Smoking cessation returns the<br />

risk to that <strong>of</strong> the general population after 20 years.<br />

H. pylori is associated with 2-6-fold increase in the risk <strong>of</strong> developing gastric<br />

cancer. In 1994, the World Health Organization designated H. pylori a group<br />

<strong>of</strong> carcinogen.<br />

Consumption <strong>of</strong> fruit, vegetables and fiber has shown in the majority <strong>of</strong> controlled<br />

studies published, a protective effect against gastric cancer. This is probably due<br />

to vitamin C or carotenes.<br />

Nitrites and nitrates found in salted, smoked and dried food lead to atrophic gastritis<br />

which in turn leads to gastric cancer.<br />

2. Genetic Factors<br />

• Small percentage <strong>of</strong> gastric cancers are hereditary known as hereditary<br />

diffuse gastric cancer (HDGC)<br />

• Blood type A<br />

• Hereditary non-polyposis colorectal cancer<br />

• E-cadherin gene mutations<br />

• A first degree relative with gastric cancer<br />

• Presently, they are not subject to preventive measures except for prophylactic<br />

gastrectomy in e-cadherin mutations<br />

Symptoms<br />

1. Early<br />

• Indigestion or burning sensation (heart burn)<br />

• Loss <strong>of</strong> appetite, especially for meat<br />

2. Late<br />

• Abdominal pain or discomfort in the upper abdomen<br />

• Nausea and vomiting<br />

• Diarrhea or constipation<br />

• Bloating <strong>of</strong> the stomach after meals<br />

• Weight loss<br />

• Weakness and fatigue<br />

• Bleeding (vomiting <strong>of</strong> blood or having blood in stool which can lead to<br />

anemia)<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 61


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

Diagnosis<br />

The most important is detailed medical history and physical examination and<br />

then to have laboratory work including full blood count since they might develop<br />

anemia secondary to blood loss<br />

• Upper GI endoscoped exam is the diagnostic method <strong>of</strong> choice. Abnormal<br />

tissue should be biopsied and sent to a pathologist.<br />

Histology<br />

There are several histological types <strong>of</strong> gastric cancer <strong>of</strong> which adenocarcinoma is<br />

by far the, the most frequent. Sarcoma and lymphoma can also incur. Two types<br />

<strong>of</strong> adenocarcinoma are recognized:<br />

1. Intestinal<br />

Resembles colon cancer, can be polypoid or ulcerated, occurs usually in the distal<br />

stomach and has prolonged precancerous phase.<br />

2. Diffuse<br />

Extends widely with no distinct margins and the glandular structure is rarely<br />

present. Patient tends to be younger and have a worse prognosis.<br />

The disease is considered early when confined to the mucosa and sub-mucosa,<br />

irrespective <strong>of</strong> lymph node status, otherwise, it is advanced.<br />

Staging<br />

• CT chest, abdomen and pelvis<br />

• Tumor marker CEA, CA<br />

Treatment<br />

It depends on the location, size and extent <strong>of</strong> disease; main therapy – surgery.<br />

Surgery<br />

Sub-total or partial gastrectomy or total gastrectomy <strong>of</strong> the stomach as well as<br />

some tissue around the stomach with basic goal <strong>of</strong> removing all cancer and a<br />

margin <strong>of</strong> normal tissue. Depending on the extent <strong>of</strong> invasion and the location<br />

<strong>of</strong> the tumor surgery may also include removal <strong>of</strong> part <strong>of</strong> the esophagus, spleen<br />

ovaries or intestine.<br />

Chemotherapy<br />

Gastric cancer is not sensitive to chemotherapy and primarily it serves to palliatively<br />

reduce the size <strong>of</strong> the tumor and increase survival time. Some <strong>of</strong> the drugs are 5-FU,<br />

BCNU, doxorubicin, cisplatin, taxotere-mitomycin. CT can be given before the<br />

surgery neoadjuvant, post-surgery adjuvant to destroy the remaining cancer cells.<br />

Radiotherapy<br />

Gastric cancer has low sensitivity to radiosensitivity. When used, it is <strong>of</strong>ten<br />

combined with surgery and chemotherapy or may be used to relieve pain or<br />

blockage by shrinking the tumor in case <strong>of</strong> terminal disease.15-16<br />

Prevention and Early Detection<br />

Gastric cancer is a very common disease that carries a high mortality. The<br />

diagnosis in early phases when better results should be expected is difficult due<br />

to the unspecificity <strong>of</strong> early symptoms.<br />

Recognition <strong>of</strong> risk factors and application <strong>of</strong> strategies directed towards their<br />

elimination are <strong>of</strong> paramount importance.<br />

Secondary prevention is the “early” detection <strong>of</strong> cancer through screening. This<br />

is done in populations where the disease is a major health problem (e.g. Japan<br />

and Costa Rica). In Japan, gas-contrast stomach fluorography is done in the mass<br />

population. Those considered abnormal (about 13%) will undergo for the studies<br />

endoscopy and biopsy.<br />

The best primary prevention strategies are:<br />

1. Smoking avoidance or cessation<br />

2. Diet rich in fruit, vegetable and fiber<br />

3. Avoidance <strong>of</strong> salted, smoked and poorly preserved food<br />

4. Eradication <strong>of</strong> H. pylori<br />

5. Mass screening is a possible strategy in high-risk populations but is not<br />

uniformly accepted.<br />

References<br />

1. Khirurgiia, S. Gastric Cancer: Current state <strong>of</strong> the problem. Part 1.<br />

Epidemiology, pathology, classifications and staging. 2007; (4):48-59 PMID:<br />

18443537 (PubMed) indexed for Mideme<br />

2. Ries LAG, Harkins D, Krapcho M, et al.: SEER Cancer Statistics Review,<br />

1975-2003. Bethesda, Md: National Cancer Institute, 2006. Also available online.<br />

Last accessed October 07, 2008.<br />

3. Qui D, Tanaka S: International comparisons <strong>of</strong> cumulative risk <strong>of</strong> stomach<br />

cancer, from Cancer Incidence in Five Continents Vol. III. Jpn J Clin Oncol 36<br />

(2):123-4, 2006.<br />

4. Stomach. In: Ries LA, Kosary CL, Hankey BF, et al., eds.: SEER Cancer Statistics<br />

Review 1973-1995. Bethesda, Md: National Cancer Institute, 1998, Section 13.<br />

5. American Cancer Society.: Cancer Facts and Figures 2005. Atlanta, Ga:<br />

American Cancer Society, 2008. Also available online. Last accessed October<br />

1, 2008.<br />

6. American Cancer Society.: Cancer Facts and Figures 2008. Atlanta, Ga:<br />

American Cancer Society, 2008. Also available online. Last accessed October<br />

1, 2008.<br />

7. Parkin DM: Global cancer statistics in the year 2000. Lancet Oncol 2 (9):<br />

533-3, 2001.<br />

8. Cancer Incidence, 2004<br />

9. Stomach. In: World Cancer Research Fund., American Institute for Cancer<br />

Research.: Food, Nutrition and the Prevention <strong>of</strong> Cancer: A Global Perspective.<br />

Washington, DC: The Institute, 1997, pp 148-175.<br />

10. Buiatti E, Palli D, Decarli A, et al.: A case-control study <strong>of</strong> gastric cancer and<br />

diet in Italy : II. <strong>Association</strong> with nutrients. Int J Cancer 45 (5): 896-901, 1990.<br />

11. Taylor PR: Prevention <strong>of</strong> gastric cancer: a miss. J Natl Cancer Inst 99 (2):<br />

101-3, 2007.<br />

12. Malila N, Taylor PR, Virtanen MJ, et al.: Effects <strong>of</strong> alpha-tocopherol and betacarotene<br />

supplementation on gastric cancer incidence in male smokers (ATBC<br />

Study, Finland). Cancer Causes Control 13 (7): 617-23, 2002.<br />

13. Wong BC, Lam SK, Wong WM, et al.: Helicobacter pylori eradication to<br />

prevent gastric cancer in a high-risk region <strong>of</strong> China: a randomized controlled<br />

trial. JAMA 291 (2): 187-94, 2004.<br />

14. Shimazu, T. Alcohol drinking and gastric cancer risk: an evaluation based<br />

on a systemic review <strong>of</strong> epidemiologic evidence among the Japanese population.<br />

Jpn J Clin Oncol 2008 38(1):8-25;doi:10.1093/jjco/hym152<br />

15. American Cancer Society. Angiogenesis Institution (National Cancer Institute)<br />

16. Cancer Treatment Options. May 2009. www.Issels.com<br />

62 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


TESTICULAR CANCER<br />

Pr<strong>of</strong> Dr. Alaa Kandil<br />

Alexandria School <strong>of</strong> Medicine, Egypt<br />

Corresponding Author: Pr<strong>of</strong>essor Dr Alaa Kandil<br />

Department <strong>of</strong> Clinical <strong>Oncology</strong> & Nuclear Medicine<br />

Alexandria School Of Medicine<br />

601 Elhoreya St, Elgawhara Bld, Zizenia, App 601<br />

E-mail: alaakandil@ hotmail.com<br />

It is estimated that 8,090 new cases <strong>of</strong> testicular cancer will be diagnosed in men,<br />

and 380 men will die <strong>of</strong> this disease in the United States in 2008.1 Testicular<br />

cancer is the most common malignancy in men aged 15 to 35 years. It accounts<br />

for approximately 1% <strong>of</strong> all cancers in men. Worldwide, testicular cancer has<br />

more than doubled in the last 40 years. Incidence varies considerably in different<br />

geographical areas, being highest in Scandinavia and Switzerland; intermediate<br />

in the United States, Australia, and the United Kingdom; and lowest in Asia and<br />

Africa. It also varies according to ethnic groups, with a much higher rate among<br />

whites than blacks in the American population.2 An annual increase <strong>of</strong> 3% is<br />

reported for Caucasian populations.3 Despite the increase in observed incidence,<br />

there has been a dramatic decrease in mortality as a result <strong>of</strong> effective treatments.<br />

Germ cell tumors (GCT) <strong>of</strong> the testis constitute 94% <strong>of</strong> testicular tumors and include<br />

five basic cell types: seminoma, embryonal carcinoma, yolk sac tumor, teratoma,<br />

and choriocarcinoma. Sixty percent <strong>of</strong> GCT are seminomas; the remainder are<br />

nonseminomatous germ cell tumors. Almost half <strong>of</strong> all germ cell tumors contain<br />

more than one <strong>of</strong> the five cell types.<br />

Three subtypes <strong>of</strong> pure seminomas have been described: classic, anaplastic, and<br />

spermatocytic. Classic seminoma accounts for 80% to 85% <strong>of</strong> all seminomas<br />

and occurs most commonly in men aged 30 to 50 years. Anaplastic seminoma<br />

accounts for 5% to 10% <strong>of</strong> all seminomas and has an age distribution similar to<br />

that <strong>of</strong> the typical subtype. A number <strong>of</strong> features suggest that anaplastic seminoma<br />

is a more aggressive and potentially more lethal variant <strong>of</strong> typical seminoma.<br />

These characteristics include greater mitotic activity, higher rate <strong>of</strong> local invasion,<br />

increased rate <strong>of</strong> metastatic spread, and higher rate <strong>of</strong> tumor marker (human<br />

chorionic gonadotropin beta, or beta hCG) production. Spermatocytic seminoma<br />

accounts for 2% to 12% <strong>of</strong> all seminomas, and nearly half occur in men older than<br />

50 years. The cells closely resemble different phases <strong>of</strong> maturing spermatogonia.<br />

The metastatic potential <strong>of</strong> this tumor is extremely low, and the prognosis is<br />

favorable.4<br />

Risk Factors<br />

Unlike most other cancers, testicular cancer is generally found in young men.5<br />

In white men, testicular cancer is the most common cancer from age 20 years to<br />

age 34 years, the second most common from age 35 years to age 39 years, and<br />

the third most common from age 15 years to age 19 years. This type <strong>of</strong> cancer is<br />

4.5 times more common among white men than black men,6 with intermediate<br />

incidence rates for Hispanics, American Indians, and Asians. High-risk groups exist.<br />

Males with cryptorchidism have 3 to 17 times the average risk. Approximately<br />

7% to 10% <strong>of</strong> patients with testicular tumors have a history <strong>of</strong> cryptorchidism.4, 7<br />

Orchiopexy may not prevent cancer in these children but allows clinical surveillance<br />

<strong>of</strong> patients with a previously impalpable gonad. There is also an increased risk<br />

in males with gonadal dysgenesis and Klinefelter syndrome.8 Men with a family<br />

history <strong>of</strong> testicular cancer may be at a higher risk <strong>of</strong> this disease.9 A history<br />

<strong>of</strong> testicular cancer is associated with a higher risk <strong>of</strong> a contralateral tumor.4, 7<br />

Although not consistently found to confer a higher risk, infertility, testicular<br />

atrophy, twinship, or abnormal semen parameters have been associated with a<br />

higher risk <strong>of</strong> testicular cancer, but the evidence is weak.7, 10<br />

An additional risk factor for the development <strong>of</strong> testicular cancer is the presence<br />

<strong>of</strong> carcinoma in situ (CIS), also called intratubular germ cell neoplasia. Testicular<br />

CIS appears to develop from fetal gonocytes and is characterized histologically<br />

by seminiferous tubules containing only Sertoli cells and malignant-appearing<br />

germ cells.<br />

If encountered in the contralateral testis, CIS is associated with the development<br />

<strong>of</strong> contralateral testicular cancer in 50% <strong>of</strong> patients at 5 years <strong>of</strong> follow-up.11<br />

CIS will be found in approximately 5% <strong>of</strong> contralateral testes (approximately the<br />

same rate as cryptorchid testes).12<br />

The association <strong>of</strong> testicular microlithiasis with testicular cancer is still <strong>of</strong><br />

questionable clinical significance.13, 14<br />

Approximately 60% <strong>of</strong> testicular cancers are localized, 24% are regional, and 14%<br />

are distant stage at diagnosis. Although there has been no appreciable change in<br />

the stage distribution at diagnosis, advances in treatment have been associated<br />

with a 60% decrease in mortality. Testicular cancer is so curable even at advanced<br />

stages and there are so few cases that it would be virtually impossible to document<br />

a decrease in mortality associated with screening.<br />

Treatment options for CIS include observation, radiation therapy, chemotherapy,<br />

and orchiectomy. Although low-dose radiation therapy can preserve Leydig cell<br />

function and prevent germ-cell tumor development, a conservative approach <strong>of</strong><br />

observation may also be warranted. Individuals at high risk (e.g., cryptorchidism,<br />

atrophic testis, and intersex conditions) require close observation.<br />

Testicular cancer survivors are at an increased risk <strong>of</strong> solid tumors for at least<br />

35 years after treatment.15 There is a low cumulative risk <strong>of</strong> metachronous<br />

contralateral testicular cancer and a favorable overall survival <strong>of</strong> patients diagnosed<br />

with metachronous contralateral testicular cancer.16<br />

Most testicular cancers are first detected by the patient, either unintentionally<br />

or by self-examination. Some are discovered by routine physical examination.<br />

However, no studies have been done to determine the effectiveness <strong>of</strong> testicular<br />

self-examination or clinical testicular examination in reducing mortality from<br />

testicular cancer. The benefit <strong>of</strong> testicular self-examination is unknown.<br />

Screening would be very unlikely to decrease mortality substantially because<br />

therapy is so effective, even for advanced stages <strong>of</strong> disease. However, early<br />

detection may have a practical impact on therapy. There is an increase in both<br />

the number <strong>of</strong> courses <strong>of</strong> chemotherapy and the extent <strong>of</strong> surgery required for<br />

treatment <strong>of</strong> advanced disease that results in higher morbidity. Patients diagnosed<br />

with localized disease require less treatment and have lower morbidity.17<br />

References<br />

1. American Cancer Society.: Cancer Facts and Figures 2008. Atlanta, Ga:<br />

American Cancer Society, 2008.<br />

2. Huyghe E, Matsuda T, Thonneau P: Increasing incidence <strong>of</strong> testicular cancer<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 63


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

worldwide: a review. J Urol 170 (1): 5-11, 2003. http://www.ncbi.nlm.nih.gov/entrez/<br />

query.fcgi?cmd=Retrieve&db=PubMed&list_uids=12796635&dopt=Abstract<br />

3. Horwich A, Shipley J, Huddart R: Testicular germ-cell cancer. Lancet 367<br />

(9512): 754-65, 2006<br />

4. Richie JP, Steele GS: Neoplasms <strong>of</strong> the testis. In: Walsh PC, Retik AB, Vaughan<br />

ED, et al., eds.: Campbell's Urology. 8th ed. Philadelphia: Saunders, 2002, pp<br />

2876-2910.<br />

5. Ries LA, Kosary CL, Hankey BF, et al., eds.: SEER Cancer Statistics Review<br />

1973-1995. Bethesda, Md: National Cancer Institute, 1998.<br />

6. Moul JW, Schanne FJ, Thompson IM, et al.: Testicular cancer in blacks.<br />

A multicenter experience. Cancer 73 (2): 388-93, 1994. http://www.<br />

ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_<br />

uids=8293405&dopt=Abstract<br />

7. Dieckmann KP, Pichlmeier U: Clinical epidemiology <strong>of</strong> testicular germ cell<br />

tumors. World J Urol 22 (1): 2-14, 2004<br />

8. Henderson BE, Benton B, Jing J, et al.: Risk factors for cancer <strong>of</strong> the testis in<br />

young men. Int J Cancer 23 (5): 598-602, 1979. http://www.ncbi.nlm.nih.gov/<br />

entrez/query.fcgi?cmd=Retrieve&db=PubMed&list_uids=37169&dopt=Abstract<br />

9. Dieckmann KP, Pichlmeier U: The prevalence <strong>of</strong> familial testicular cancer:<br />

an analysis <strong>of</strong> two patient populations and a review <strong>of</strong> the literature. Cancer 80<br />

(10): 1954-60, 1997<br />

10. Jacobsen R, Bost<strong>of</strong>te E, Engholm G, et al.: Risk <strong>of</strong> testicular cancer in men<br />

with abnormal semen characteristics: cohort study. BMJ 321 (7264): 789-92, 2000<br />

11. Jørgensen N, Müller J, Giwercman A, et al.: Clinical and biological significance<br />

<strong>of</strong> carcinoma in situ <strong>of</strong> the testis. Cancer Surv 9 (2): 287-302, 1990<br />

12. Dieckmann KP, Loy V: Prevalence <strong>of</strong> contralateral testicular intraepithelial<br />

neoplasia in patients with testicular germ cell neoplasms. J Clin Oncol 14 (12):<br />

3126-32, 1996<br />

13. Holm M, Hoei-Hansen CE, Rajpert-De Meyts E, et al.: Increased risk <strong>of</strong><br />

carcinoma in situ in patients with testicular germ cell cancer with ultrasonic<br />

microlithiasis in the contralateral testicle. J Urol 170 (4 Pt 1): 1163-7, 2003<br />

14. Rowland RG: Editorial: testicular malignancies--high chance <strong>of</strong> curecontinuing<br />

opportunities to refine treatment. J Urol 170 (4 Pt 1): 1168, 2003<br />

15. Travis LB, Fosså SD, Schonfeld SJ, et al.: Second cancers among 40,576<br />

testicular cancer patients: focus on long-term survivors. J Natl Cancer Inst 97<br />

(18): 1354-65, 2005<br />

16. Fosså SD, Chen J, Schonfeld SJ, et al.: Risk <strong>of</strong> contralateral testicular cancer:<br />

a population-based study <strong>of</strong> 29,515 U.S. men. J Natl Cancer Inst 97 (14): 1056-<br />

66, 2005<br />

17. Sagalowsky AI: Expectant management <strong>of</strong> stage A nonseminomatous testicular<br />

tumors. In: Ratiff TL, Catalona WJ, eds.: Genitourinary Cancer. Boston: Martinus<br />

Nijh<strong>of</strong>f, 1987, pp 225-237.<br />

THYROID CANCER EPIDEMIOLOGY AND PREVENTION<br />

Saad Al Shehri, MD, FRCSC, FACS<br />

King Abdulaziz <strong>Medical</strong> City for National Guard, Riyadh, KSA<br />

Corresponding Author: Saad Al Shehri, MD, FRCSC, FACS<br />

Department <strong>of</strong> Surgery, P.O. Box 22490, Riyadh 11426<br />

Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

E-mail: shehris@ngha.med.sa<br />

Introduction<br />

The thyroid gland is made up <strong>of</strong> two lateral lobes which extend from the sides <strong>of</strong><br />

the thyroid cartilage to the sixth tracheal ring. These are joined together by the<br />

isthmus which overlies the second to fourth tracheal rings.<br />

The thyroid gland is the largest organ specialized in endocrine function in the<br />

human body.<br />

Cells <strong>of</strong> Origin:<br />

Thyroid gland contains two embryologically and functionally different cell types:<br />

• The neuroendocrine Calcitonin producing C – cell and<br />

• Endodermally derived follicular cell that produces T4 and Thyroglobulin<br />

Thyroid cancers are derived from these two embryonic cell lines.<br />

Classification <strong>of</strong> Thyroid Cancer:<br />

1. Papillary Carcinoma<br />

• 80% <strong>of</strong> thyroid cancer<br />

• Peak age: 3rd, 4th decades<br />

• More in women (3 folds)<br />

• More in white<br />

2. Follicular Carcinoma<br />

• 10% <strong>of</strong> thyroid cancer<br />

• Peak age: 4th and 5th decades<br />

• More frequent in women<br />

• Uncommon in black<br />

3. Medullary Thyroid Carcinma (MTC)<br />

• 5 – 7% <strong>of</strong> thyroid cancer<br />

• Sporadic type 80%<br />

• Familial type 20%, point <strong>of</strong> mutation in the RET proto – oncogene. In kindred<br />

with MTC can identify affected persons can be identified before clinical or<br />

chemical changes can be detected (1,2)<br />

4. Anaplastic Thyroid Carcinoma<br />

• Arises from less differentiated thyroid cell<br />

• Caused by external radiation (3)<br />

• Occurs with greater frequency in areas <strong>of</strong> low iodine diet and endemic<br />

goiter area (4)<br />

• More in elderly, 76% more in women (ratio 2:1)<br />

• 1/3 <strong>of</strong> specimen have areas <strong>of</strong> well differentiated, rare in children<br />

• Treatment ranges from debulking and post operatively radiation to systemic<br />

chemotherapy<br />

• Median survival is 6 months<br />

64 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


5. Thyroid Lymphoma<br />

• It occurs with greater frequency than anaplastic carcinoma<br />

• Incidence has increased in recent years<br />

• Hashimoto’s disease increased patient’s risk <strong>of</strong> thyroid lymphoma about<br />

70 fold (5)<br />

• Mean: age from 60 to 65<br />

• Female to Male: 8:1 (6)<br />

• Prognosis depends on stage if confined to thyroid, 5 year survival rate is 85%<br />

if extending into surroundings neck structure (35%) and for disseminated<br />

disease rate (5%) (7)<br />

• Treatment is radiotherapy alone and surgery and radiotherapy together are<br />

highly effective<br />

6. Secondary Thyroid Tumor<br />

• Microscopic metastasis to thyroid gland found in autopsy specimens from<br />

patients with malignant melanoma, lung and breast carcinoma<br />

• Usually undetectable clinically<br />

• Does not alter the thyroid function<br />

Epidemiology<br />

Thyroid cancer is a relatively uncommon clinical problem. In USA, account for<br />

less than 1.5 <strong>of</strong> all cancer cases. This appears to be steadily increasing in incidence.<br />

In the 1970’s, 8000 new cases / year. In 2002, there were 20, 700 cases /year but<br />

the death rate remained the same with fewer than 1300 case / year.<br />

Thyroid cancer constitutes 92% <strong>of</strong> endocrine cancers (8). The incidence <strong>of</strong> thyroid<br />

cancer is high in males with thyroid nodules and among patients younger than 30<br />

years old or older than 60 years <strong>of</strong> age (9)<br />

The prevalence <strong>of</strong> occult thyroid carcinoma in autopsy specimen is about 10%.<br />

(10,11)<br />

In Saudi <strong>Arab</strong>ia(12):<br />

There were 415 cases <strong>of</strong> thyroid cancer accounting for 6% <strong>of</strong> all newly diagnosed<br />

cases in year 2004. This cancer ranked second among female population and 14th<br />

among male population. It affected 87 (21%) males and 328 (79%) females with<br />

a male to female ratio <strong>of</strong> 100:266.<br />

Risk Factors<br />

1. Benign Thyroid Disease<br />

Because thyroid cancer occurs more <strong>of</strong>ten in solitary nodules than in multinodular<br />

goiters (MNG) the two are <strong>of</strong>ten managed differently; however, this may lead to<br />

a serious diagnostic error. Up to 50% <strong>of</strong> thyroid glands, with palpable solitary<br />

nodules have multiple lesions by ultrasound or at surgery. (13,14)<br />

A dominant hyp<strong>of</strong>unctional nodule in MNG or a diffuse goiter with large palpable<br />

cervical lymph nodes should be evaluated for carcinoma. Hashimato’s thyroiditis<br />

can transform to lymphoma in 20 – 30% <strong>of</strong> the cases.<br />

2. Radiation Exposure<br />

This is an important risk factor for thyroid carcinoma and other head and neck<br />

neoplasms (15,16,17). <strong>Medical</strong> workers exposed to radiation have a significantly<br />

high prevalence <strong>of</strong> thyroid cancer than controls (18).<br />

The risk <strong>of</strong> developing carcinoma is greatest after radiation exposure in childhood.<br />

Likewise, persons exposed to radiation from atomic weapons and nuclear fallout<br />

accidents have a highly incidence <strong>of</strong> thyroid cancer.<br />

The latest period for developing abnormality after radiation exposure is commonly<br />

between 10 – 20 years <strong>of</strong> age, but it can happen during childhood.<br />

3. Diet<br />

Epidemiologic studies relating diet, iodine consumption and iodine deficiency to<br />

thyroid cancer have not clearly demonstrated a universal link.<br />

4. Genetic Factors<br />

The evidence is growing that genetic factors may play a role in a small percentage <strong>of</strong><br />

papillary and follicular thyroid carcinomas. Good example <strong>of</strong> that the well known<br />

association between Gardner’s Syndrome (familial polyposis) and Cawden’s<br />

disease (familial goiter and skin hematomas). (19,20)<br />

5. Hereditary Medullary Thyroid Carcinoma<br />

• A manifestation <strong>of</strong> multiple endocrine neoplasia:<br />

• MEN 2A: Medullary thyroid cancer, pheochromocytoma,<br />

parathyroid neoplasia<br />

• MEN 2B: Medullary thyroid cancer, Marfanoid features<br />

• Familial medullary thyroid carcinoma with no other endocrine diseases.<br />

Presenting Signs and Symptoms<br />

Patients with thyroid cancer may have one or more <strong>of</strong> the following signs and<br />

symptoms:<br />

1. Firm hard, painless lump in the front <strong>of</strong> the neck<br />

2. Swelling in the neck (goiter)<br />

3. Swallowing difficulty (pressure)<br />

4. Hoarseness or change in voice (pressure or invasion <strong>of</strong> recurrant laryngeal nerve)<br />

5. Coughing with hemoptysis (invasion <strong>of</strong> trachea)<br />

6. Breathing difficulty (pressure)<br />

7. Unexplained bone fractures (follicular)<br />

8. Severe flushing or diarrhea (medullary)<br />

Screening<br />

Genetic testing can identify members <strong>of</strong> an affected kindred at birth. (21) It is<br />

indicated if a patient with familial MTC has a mutation in the RET proto – oncogene<br />

and is then indicated in all first – degree relatives. Those with a positive test,<br />

including children, should undergo thyroidectomy. Serum calcitonine measurement<br />

should be done during follow up visits. Patients with MEN 2A and MEN 2B<br />

should have urinary catechalamines and metanephrines and serum calcium level<br />

during follow up.<br />

Although the molecular abnormalities in papillary and follicular thyroid carcinoma<br />

are undoubtedly important, no direct clinical application at this time, but it might<br />

in the future.<br />

Prognosis and Outcome<br />

For Well Differentiated Thyroid Cancer<br />

Patient under 45 years old have better prognosis, women do better than men. There<br />

is linear relationship between tumor size and prognosis, also histological type and<br />

extent or local invasion <strong>of</strong> tumor are significant prognostic factors.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 65


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

For Medullary Thyroid Cancer<br />

Early therapy has improved survival with MTC in patients with MEN 2A, and<br />

has decreased the frequency <strong>of</strong> distant metastasis. The prognosis <strong>of</strong> patients<br />

with the MEN 2B is substantially worse than patients with MEN 2A but it is also<br />

improved by early diagnosis and therapy. Outcome is most favorable in patients<br />

with familial MTC without associated endocrine tumors.<br />

Anaplastic Tumors<br />

Dismal prognosis with a median survival <strong>of</strong> 6 – 12 months.<br />

Prevention<br />

Primary: Eliminate the Risk Factors<br />

Due to the fact that not all the risk factors for thyroid malignancy are known, it<br />

is difficult to conduct complete primary prevention.<br />

Factors like exposure to radiation can be controlled by minimizing x – ray exposure<br />

to children and thyroid gland by using shields.<br />

There is a scientific and political responsibility to control nuclear plants and avoid<br />

accidents, and early public notification if occurred.<br />

Secondary: Early Detection and Treatment<br />

In case <strong>of</strong> familial thyroid carcinoma:<br />

• To conduct genetic screening<br />

• And regular check up by physical exam and ultrasound testing. If any lesion<br />

is noted in thyroid gland, obtain biopsy.<br />

Treatment includes:<br />

• Surgical resection<br />

• Clearance <strong>of</strong> lymph nodes, if any<br />

• + Radioactive Iodine<br />

• + External beam radiation<br />

• Also thyroids hormone supplements post operatively<br />

Tertiary: Prevent Recurrence and Decrease Mortality<br />

For Well Differentiated Thyroid Carcinoma: Testing for metastasis using thyroid<br />

scan and also monitoring the serum thyro globuline level.<br />

For Medullary Thyroid Carcinoma: The monitoring <strong>of</strong> serum calcitonin gives<br />

very good indicator <strong>of</strong> treatment, efficacy and outcome.<br />

In advanced thyroid cancer, palliative measures including pain control, protect<br />

airway (Tracheostomy), maintains enteral feeding (Gastrostomy).<br />

Recommendations <strong>of</strong> Prevention and Early Detection <strong>of</strong> Thyroid Cancer<br />

Primary Preventions:<br />

• Avoid exposure to radiation neither accidental nor excessive medical<br />

exposure.<br />

• Although it is not clear risk factor, but it is advisable to avoid low iodine<br />

in diet<br />

Early Detection:<br />

• No clinical and cost effective method with proven benefit for screening the<br />

population for thyroid cancer in general.<br />

Early Detection in Special Population:<br />

• Calcitonin level in patient with MEN Syndrome for medullary carcinoma<br />

• Close observation for certain benign condition e.g. Hashimoto’s disease,<br />

Gardner’s Syndrome and Cawden’s Disease since there is association with<br />

thyroid cancer.<br />

The serum calcitonin and serum thyroglobulin can be used as a marker for early<br />

detection <strong>of</strong> recurrence <strong>of</strong> some <strong>of</strong> the thyroid cancers.<br />

References<br />

1. Sondik EJ and other: 1986 Annual Cancer Statistics Review. Belhesda 1986.<br />

Department <strong>of</strong> Health and Human Services. Public Health Service. National<br />

Cancer Institute.<br />

2. Cornea P, Chen VW: Endocrine Gland Cancer, Cancer 75: 338, 1995.<br />

3. Belfiore A and others: Cancer Risk in Patients with Cold Thyroid Nodules:<br />

Relevance <strong>of</strong> Iodine Intake, Sex, Age and Multinodulanity, A.M.J. Med 93: 363,<br />

1992.<br />

4. De Vathaire F and others: Epidemiological Evidence for a Common Mechanism<br />

for Neuroblastoma and Differentiated Thyroid Tumor, BrJ. Cancer 65: 425, 1992.<br />

5. Mazzferri El, de los Santos ET, R<strong>of</strong>agha – Keyhanis: Solitary Thyroid Nodules:<br />

Diagnosis and Management, Med Chin North Am 72: 1177, 1988.<br />

6. Al – Saleh Ms., Al – Kattan KM: Incidence <strong>of</strong> Carcinoma in multinodular<br />

Goiter in Saudi <strong>Arab</strong>ia, J.R. Coll Surg. Edinb 39: 106, 1994.<br />

7. Roher HD: Thyroid Cancer: is the Radical Surgical Principle Changing?<br />

Longerbecks Arch Chir 370:1; 1987.<br />

8. Schneider AB and others: Radiation Induced Tumors <strong>of</strong> the Head and Neck<br />

Following Childhood Immediation: Prospective Studies. Medicine (Baltimore)<br />

64: 1, 1985.<br />

9. Schneider AB and others: Radiation – Induced Thyroid Carcinoma: Clinical<br />

Course and Results <strong>of</strong> Therapy in 296 Patients. Ann Intern Med 105: 405, 1986.<br />

10. Valdiserri RO, Borochovitz D: Histologic Changes in Previously Innadiated<br />

Thyroid Gland:. Arch Path Lab Med 104: 150, 1980<br />

11. Antonelli A and others: Risk <strong>of</strong> Thyroid Nodules in Subjects Occupationally<br />

Exposed to Radiation: A Cross – Sectional Study, Occup Envison Med 52: 500,<br />

1995.<br />

12. Donis – Keller H and others: Mutations in the RET Proto – Oncogene are<br />

Associated with Menza and FMTC, Hum Genet 2: 852, 1993.<br />

13. Mulligan LM and Others: Germline Mutations <strong>of</strong> the RET Proto Oncogene<br />

in Multiple Endocrine Neoplasia Type 2A, Nature 363: 458, 1993.<br />

14. Plial RO, Bussey HJR, Thomson JPS: Adenomatous Polyposis An <strong>Association</strong><br />

with Carcinoma <strong>of</strong> the Thyroid Br. J. Surg 74: 377, 1987.<br />

15. Sogol PB, Sugawara M, Gordon HE, et al: Cowden’s Disease: Familial Goiter<br />

and Skin Hamartomas. A Report <strong>of</strong> Three Cases. West J Med 139: 324, 1983.<br />

16. Saudi Cancer Registry, 2004<br />

17. Komorowski RA, Hanson GA, Garancis JC: Anaplastic thyroid carcinoma<br />

following low – dose irradiation, Am J Clin Pathol 70: 303, 1978.<br />

18. Hedinger C: Geographic pathology <strong>of</strong> thyroid diseases. Pathol Res Pract<br />

171:285. 1981.<br />

19. Holm LE, Blomgren H. Lowenhagen T: Cancer risks in patients with chronic<br />

lymphocytic thyroiditis. N Engl J Med 312: 601. 1985.<br />

20. Mazzaferri EL, Oertel YC: Primary malignant lymphoma and related<br />

lymphoproliferative disorders. In Mazzaferri EL, Samaan N, editors:<br />

21. Hamburger JI, Miller JM, Kini SR: Lymphoma <strong>of</strong> the thyroid, Ann Intern<br />

Med 99: 685, 1983.<br />

66 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


COLORECTAL CANCER:<br />

PREVENTION AND EARLY DETECTION<br />

Jazieh, A., Abulkhair, O., Olayan, A, Mahmood, R.<br />

King Abdulaziz <strong>Medical</strong> City, Riyadh, KSA<br />

Corresponding Author: Abdul Rahman Jazieh, MD, MPH<br />

Chairman, Department <strong>of</strong> <strong>Oncology</strong>, King Abdulaziz <strong>Medical</strong> City<br />

P.O. Box 22490 Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

E-mail: jazieha@ngha.med.sa<br />

Abstract<br />

Colorectal cancer (CRC) is the third most common malignancy and second leading<br />

cause <strong>of</strong> cancer death irrespective <strong>of</strong> gender.<br />

A number <strong>of</strong> genetic, environmental and life style risk factors are involved<br />

in the pathogenesis <strong>of</strong> CRC. In addition, several studies have established a<br />

number <strong>of</strong> factors with a protective role against development <strong>of</strong> colorectal cancer.<br />

Identification <strong>of</strong> these factors and subsequent modification can help us in preventing<br />

this potentially lethal disease.<br />

Screening for CRC can help us in two ways. First, it can detect sub-clinical<br />

abnormalities which may have a potential for malignant transformation and prompt<br />

us for early action and thus can prevent CRC. Secondly, it helps in the detection<br />

<strong>of</strong> CRC at an early stage when clinical symptoms may not be that evident for an<br />

affected individual to seek medical advice. However, treatment at this early stage<br />

is most effective and curative in most cases.<br />

The following manuscript describes the summary <strong>of</strong> protective and risk factors <strong>of</strong><br />

CRC and screening modalities which can help us in prevention and early detection<br />

<strong>of</strong> colorectal cancer.<br />

Introduction<br />

Colorectal cancer (CRC) is the third most common malignancy and potentially<br />

a lethal disease if not treated.¹ It is the second leading cause <strong>of</strong> cancer death in<br />

the US.² The importance <strong>of</strong> prevention and early detection <strong>of</strong> certain cancers<br />

cannot be over-emphasized due to the fact that many malignancies are potentially<br />

preventable. 3, 4<br />

Epidemiology<br />

The number <strong>of</strong> diagnosed cases <strong>of</strong> CRC annually in the U.S. is close to 150,000<br />

and approximately one third <strong>of</strong> them die from the diseas.2<br />

CRC is seen mostly after the age <strong>of</strong> 40, with the risk increasing with each decade.<br />

5,6 90 percent <strong>of</strong> cases are seen in age more than 50 years. It is seen most common<br />

in North America, Australia and Europe as compared to Africa and Asia.7<br />

The overall 5-year survival rate for colorectal cancer is approximately 66 percent.<br />

Although there have been advances in the surgical and adjuvant treatment <strong>of</strong> CRC,<br />

the prognosis remains poor for patients who present at an advanced stage. 8, 9<br />

Pathogenesis <strong>of</strong> Colorectal Cancer<br />

Adenomatous polyps are thought to be a precursor <strong>of</strong> CRC. 10 It takes about 10<br />

years in the progression <strong>of</strong> a polyp from a small size to a large one, then dysplasia<br />

and ultimately neoplastic appearance. 11, 12<br />

This has led to the development <strong>of</strong> screening tools for early detection and removal<br />

<strong>of</strong> adenomatous polyps with evidence <strong>of</strong> lower incidence <strong>of</strong> CRC in such patients.13<br />

Risk Factors for Colorectal Cancer<br />

The various risk factors increasing the likelihood <strong>of</strong> development <strong>of</strong> colorectal<br />

cancer are listed in Table 1.<br />

A. Familial Adenomatous Polyposis (FAP)<br />

FAP is caused by a mutation in adenomatous polyposis coli (APC) gene and<br />

accounts for less than 1% <strong>of</strong> cases <strong>of</strong> CRC.14, 15 It is characterized by appearance<br />

<strong>of</strong> numerous colonic polyps appearing in childhood and may transform into<br />

malignancy by age 45 in 90% untreated cases.<br />

B. Hereditary nonpolyposis colorectal cancer (HNPCC)<br />

Also know as Lynch syndrome.16 It accounts for 1-5% <strong>of</strong> CRC. It is an autosomal<br />

dominant syndrome; caused by mutations in mismatch repair genes; with early age<br />

<strong>of</strong> onset and involvement <strong>of</strong> proximal colon. It may also involved extra colonic<br />

sites including endometrial carcinoma, stomach, small bowel, Hepatobiliary system.<br />

C. Personal history <strong>of</strong> sporadic cancers or adenomatous polyps<br />

1.5 to 3% patients with a history <strong>of</strong> resected colorectal cancer can develop CRC<br />

in first 5 years. Presence <strong>of</strong> adenomas also poses a threat for CRC especially if<br />

multiple polyps are present.17<br />

D. Family history <strong>of</strong> sporadic cancers or adenomatous polyps<br />

A first degree relative with CRC increases the risk by 1.7 fold with further increase<br />

if two first degree relatives are involved. The risk is also present if there is a family<br />

history <strong>of</strong> a polyp larger than 1 cm in size.18<br />

E. Inflammatory bowel disease<br />

Inflammatory bowel disease (ulcerative colitis and Crohn’s disease) also increase<br />

the risk <strong>of</strong> CRC by 5 to 15-fold. The risk is increased at 8 to 10 years after the<br />

onset <strong>of</strong> symptoms. 19-21<br />

F. Other risk factors<br />

Various other risk factors have been identified as risk factors for development <strong>of</strong><br />

colorectal cancer. These include:<br />

• Diabetes mellitus and insulin resistance22-27<br />

• Cholecystectomy28-32<br />

• Alcohol33-39<br />

• Obesity40-43<br />

• Coronary heart disease44<br />

• Cigarette smoking45, 46<br />

• Acromegaly47, 48<br />

• Red meat and caffeine49-55<br />

• History <strong>of</strong> radiation therapy for prostate cancer56<br />

• HIV57, 58<br />

• Prior treatment for Hodgkin’s Lymphoma59<br />

Protective Factors for Colorectal Cancer<br />

A number <strong>of</strong> protective factors have been identified and have been summarized<br />

in Table 2.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 67


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

A. Diet<br />

The dietary factors with a protective role against colorectal cancer include:<br />

• High Fiber diet, diet high in fruits and vegetables60-63<br />

• Folic acid97, 98<br />

• VitaminB6 64<br />

• Calcium65-72<br />

• Magnesium73<br />

B. Non Steroidal Anti Inflammatory Drugs (NSAIDs)<br />

There is evidence on bases <strong>of</strong> observational studies that aspirin and NSAIDs<br />

(Celecoxib) may have a protective role against development <strong>of</strong> colorectal cancer,<br />

however, there is also documented harm <strong>of</strong> use <strong>of</strong> aspirin and NSAIDs due to<br />

their side effects.<br />

C. Physical Activity<br />

Regular physical activity has been identified as a protective factor. Several studies<br />

support this finding.76-78<br />

D. Chemoprevention<br />

D, L, alpha-difluoromethylornithine (DFMO) and sulindac (a NSAID) used in a<br />

group <strong>of</strong> patients with history <strong>of</strong> colonic adenoma showed reduction in development<br />

<strong>of</strong> recurrent adenomas in a clinical trial.79 However, further investigation is<br />

required to confirm the benefits <strong>of</strong> this in the colon cancer prevention.<br />

E. Other Protective Factors/Agents<br />

Other protective factors have been reported which includes:<br />

• Hormone replacement therapy8085 (Not routinely recommended for<br />

prevention <strong>of</strong> colorectal cancer in women due to long-term, risks)86<br />

• Statins87-89<br />

• Antioxidants90<br />

• Omega 3 Fatty Acids (Fish Oil)91<br />

• Garlic92, 93<br />

F. Screening for Colorectal Cancer<br />

Screening for colorectal cancer is summarized in Table 3. (95-101)<br />

Screening is divided into 3 population groups:<br />

1. Population at Average Risk<br />

• Age more than or equal to 50 years<br />

• No history <strong>of</strong> adenoma<br />

• No history <strong>of</strong> inflammatory bowel disease<br />

• Negative family history<br />

2. Population at Increased Risk<br />

• Personal history <strong>of</strong> adenoma<br />

• Personal history <strong>of</strong> colorectal cancer<br />

• Personal history <strong>of</strong> inflammatory bowel disease<br />

• Positive family history <strong>of</strong> first degree relative with colorectal cancer or<br />

adenoma.<br />

3. Population with Hereditary Risk<br />

• Hereditary non-polyposis colorectal cancer syndrome (HNPCC)<br />

• Familial Adenomatous Polyposis (FAP)<br />

G. Tests Used for Screening for Colorectal Cancer<br />

Screening with Stool-Based Tests<br />

• Guaiac-based fecal occult blood test. (Gfobt)<br />

• Immunochemical tests for fecal blood<br />

• Fecal DNA tests<br />

Colon Imaging and Direct Visualization<br />

• Double contrast barium enema. (DCBE)<br />

• Sigmoidoscopy<br />

• Colonoscopy<br />

• Computed tomographic colonography. (CTC)<br />

H. Recommendations for Screening for Average Risk Population<br />

• Offer screening beginning at age 50 years for average risk patients.<br />

Colonoscopic detection <strong>of</strong> colorectal cancer is uncommon in asymptomatic<br />

people under the age <strong>of</strong> 50 [94].<br />

• Discontinue screening when the individual’s estimated life expectancy is<br />

less than 10 years.<br />

• No single test is <strong>of</strong> unequivocal superiority. Incorporating patient personal<br />

preferences may increase the likelihood that screening will occur.<br />

• Screening should be supported by a program that assures proper follow-up<br />

<strong>of</strong> abnormal findings, and ongoing testing at identified intervals.<br />

I. Recommendations for Screening for High Risk Population<br />

• People with a first-degree relative (parent, sibling, or child) with colon<br />

cancer or adenomatous polyp diagnosed at age less than 60 years, or two<br />

first-degree relatives diagnosed at any age should be advised to have screening<br />

colonoscopy starting at age 40 years, or 10 years younger than the earliest<br />

diagnosis in their family, whichever comes first, and repeated every 5 years.<br />

• People with a first-degree relative with colorectal cancer or adenoma<br />

diagnosed at age more than 60 years should be advised to have screening<br />

as average-risk persons but beginning at age 40 years.<br />

• People with two or more second-degree relatives (grandparent, aunt, uncle)<br />

with colorectal cancer should similarly be advised to begin screening at<br />

age 40 years.<br />

• People with one second-degree relative or third-degree relative (greatgrandparent<br />

or cousin) with colorectal cancer should be advised to be<br />

screened as average risk persons.<br />

• Individual at high risk with familial syndromes (NHPCC, FAP) should be<br />

screened for CRC with colonoscopy at frequent specified intervals.<br />

68 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


Table 1: Risk Factors for Colorectal Cancer<br />

Risk Factors Ralative Risk Age Comments<br />

Familial adenomatous polyposis<br />

(FAP)<br />

Hereditary nonpolyposis colorectal<br />

cancer (HNPCC)<br />

Personal history <strong>of</strong> colorectal cancer<br />

or adenomatous polyps<br />

Family history <strong>of</strong> colorectal cancer or<br />

adenomatous polyps<br />

- 90% absolute risk <strong>of</strong> development<br />

<strong>of</strong> colorectal cancer by age 45 in<br />

mutation carriers<br />

- 80% absolute risk <strong>of</strong> development<br />

<strong>of</strong> colorectal cancer by age 75 in<br />

mutation carriers<br />

3.5 to 6.5 with adenomatous polyps<br />

>1cm<br />

1.7 fold increased risk with single<br />

affected first degree relative with<br />

CRC<br />

Inflammatory bowel disease 5 to 15-fold increased risk with<br />

pancolitis. Three-fold relative risk<br />

with left-sided disease<br />

Diabetes mellitus CRC 30% higher in diabetics.<br />

Relative Risk 1.30<br />

- Appearing during childhood<br />

- Symptoms at 16 years<br />

- Colorectal cancer by age 45 (in 90%<br />

<strong>of</strong> untreated individuals)<br />

- Early age <strong>of</strong> onset (mean age 48<br />

years, some present in 20s)<br />

- Increased risk <strong>of</strong> index case<br />

diagnosed at age 1cm<br />

has same significance as positive<br />

family history <strong>of</strong> CRC<br />

- Increased risk <strong>of</strong> ulcerative colitis<br />

and primary sclerosing cholangistis<br />

are together<br />

Cholecystectomy Standardized incidence ratio 1.16 - Data shows slightly increased risk <strong>of</strong><br />

right-sided colon cancer<br />

- Conflicting data also reported<br />

Alcohol Consumption Adjusted relative risk 1.41 - Seen with alcohol consumption ><br />

45 g/day<br />

Obesity 1.5-fold increased risk - Increased likelihood <strong>of</strong> death from<br />

CRC<br />

Cigarette Smoking Relative risk <strong>of</strong> 1.18 for CRC<br />

development.<br />

Relative risk <strong>of</strong> 1.25 for death from<br />

CRC<br />

- Stronger association with rectal<br />

cancer<br />

- Stronger risk for development <strong>of</strong><br />

polyps<br />

Coronary heart disease - Increased risk <strong>of</strong> advanced<br />

adenomas<br />

Acromegaly - Increase prevalence <strong>of</strong> polyps in<br />

males only<br />

Red meat and caffeine - Increased risk <strong>of</strong> CRC with longterm<br />

red meat/ processed meat<br />

consumption<br />

- Data inconsistent and controversial<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 69


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

Table 2: Protective Factors for Colorectal Cancer<br />

Protective Factors Relative Risk/Protective Effect Comments<br />

Dietary Factors<br />

Diet high in fruits and vegetables<br />

Diet low in red meat/cholesterol Relative risk <strong>of</strong> 3.26 with high cholesterol<br />

Fiber Up to 40% reduction in CRC risk as claimed in one<br />

study (on doubling fiber intake in population with<br />

low fiber intake)<br />

0.5 - Protective effect seen in may studies<br />

- Discordant data also published<br />

- Many conflicting studies<br />

- Protective role remains controversial<br />

Folic Acid 0.25 - Protective effect seen after 15 years use<br />

- Studies suggest protective effect seen with dietary<br />

folic acid<br />

Vitamin B6 (Pyridoxine) 0.42 - Total vitamin B6 intake inversely related with<br />

colon cancer risk<br />

Calcium 0.65 (Distal Colon Cancers) - Higher calcium intake > 1250mg/d protective<br />

Magnesium 0.59 - Up to 40% risk reduction observed<br />

Omega 3 fatty acids (mainly fish oil) 0.88 - Protective effect with high consumption <strong>of</strong> fish<br />

Garlic - Reduced risk <strong>of</strong> colonic adenomas<br />

PHYSICAL ACTIVITY 0.60 (light occupational activity)<br />

0.45 (moderate/heavy activity)<br />

0.33 (most active men)<br />

ASPIRIN and NSAIDS - No adequate studies available - Observational<br />

and intervention trial evidence supports protective<br />

effect<br />

CHEMOPREVENTION (Combination <strong>of</strong> D,L<br />

alpha – diflouromethylornithine DFMO and<br />

sulindac (NSAID)<br />

70% reduction in recurrent adenomas<br />

90% reduction in advanced adenomas<br />

95% reduction in multiple adenomas<br />

HORMONE REPLACEMENT THERAPY Risk reduction by 40% with combined estrogen<br />

plus progestin.<br />

- Protective effect in patients with history <strong>of</strong><br />

colonic adenoma<br />

- Controversial data about protective effect.<br />

- Not routinely recommended due to associated<br />

risks<br />

STATINS - Conflicting data about protective effect<br />

- Use <strong>of</strong> statins for 5 years proved protective<br />

Negative family<br />

history<br />

Screening Only<br />

Table 3: Screening for Colorectal Cancer<br />

Population at Risk Intervention Procedure Frequency Start Age (Yrs) Stop Age (Yrs) Further Action<br />

AVERAGE RISK<br />

Colonoscopy Every 10 yrs 50 70 Polypectomy if<br />

polyps found<br />

Age more than or<br />

equal to 50 years<br />

No history <strong>of</strong><br />

adenoma<br />

Screening and<br />

Prevention<br />

Computed<br />

Tomographic<br />

Colonography<br />

Flexible<br />

Sigmoidoscopy<br />

Every 5 yrs<br />

Every 5 yrs<br />

50<br />

50<br />

70<br />

70<br />

Colonoscopy if any<br />

positive findings<br />

No history <strong>of</strong><br />

Double Contrast Every 5 yrs 50 70<br />

inflammatory bowel<br />

Barium Enema<br />

disease<br />

Guaiac Based Fecal<br />

Occult Blood Testing<br />

Annually<br />

50<br />

70<br />

Fecal<br />

Immunohistochemical Annually<br />

Based Testing for<br />

Blood<br />

Stool DNA Testing Uncertain<br />

Frequency<br />

70 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info<br />

50<br />

70<br />

50 70<br />

Flexible<br />

sigmoidoscopy or<br />

CT Colonography if<br />

positive


Population at Risk Intervention Procedure Frequency Start Age (Yrs) Stop Age (Yrs) Further Action<br />

INCREASED RISK<br />

Within 5 years <strong>of</strong><br />

detection <strong>of</strong> polyp<br />

70 Polypectomy<br />

Personal history <strong>of</strong><br />

adenoma, colorectal<br />

or inflammatory<br />

bowel disease IBD<br />

(Crohn’s disease or<br />

Ulcerative colitis)<br />

Positive family<br />

history <strong>of</strong> first<br />

degree relative with<br />

colorectal cancer <strong>of</strong><br />

adenoma<br />

HEREDITARY<br />

RISK<br />

Non-Polyposis<br />

Colorectal Cancer<br />

(HNPCC)/Lynch<br />

Syndrome<br />

Screening Colonoscopy<br />

Within 1 yr <strong>of</strong><br />

personal history <strong>of</strong><br />

CRC<br />

Every 1 – 2 years<br />

(for symptomatic<br />

IBD)<br />

Screening Colonoscopy Every 5 years<br />

When detected post<br />

operative<br />

70 Repeat every 3 yrs if<br />

normal<br />

8-10 years after IBD<br />

symptoms Biopsies<br />

At age 40 yrs or 10<br />

yrs prior to earliest<br />

cancer in family<br />

Screening Colonoscopy Every 1-2 years 20-25 or 10 years<br />

prior<br />

References<br />

1. Shike M, Winawer SJ, Greenwald PH, et al.: Primary prevention <strong>of</strong> colorectal<br />

cancer. The WHO Collaborating Centre for the Prevention <strong>of</strong> Colorectal Cancer.<br />

Bull World Health Organ 68 (3): 377-85, 1990<br />

2. American Cancer Society.: Cancer Facts and Figures 2008. Atlanta, Ga:<br />

American Cancer Society, 2008<br />

3. Murray, C, Lopez, A. The Global burden <strong>of</strong> disease. World Health Organization,<br />

Geneva 1996<br />

4. Danaei, G, Vander Hoorn, S, Lopez, AD, et al. Causes <strong>of</strong> cancer in the world:<br />

comparative risk assessment <strong>of</strong> nine behavioral and environmental risk factors.<br />

Lancet 2005; 366:1784<br />

5. Eddy, DM. Screening for colorectal cancer. An Intern Med 1990; 113:373<br />

6. Jemal, A, Siegel, R, Ward, E, et al. Cancer statistics, 2008. CA Cancer J Clin<br />

2008; 58:71<br />

7. Parkin, DM, Pisani, P, Ferlay, J Globa; Cancer statistics. CA Cancer J Clin 1999; 49:33<br />

8. Moertel CG, Fleming TR, Macdonald JS, et al.: Levamisole and fluorouracil for<br />

adjuvant therapy <strong>of</strong> resected colon carcinoma. N Engl J Med 322 (6): 352-8, 1990<br />

9. Krook JE, Moertel CG, Gunderson LL, et al.: Effective surgical adjuvant therapy<br />

for high-risk rectal carcinoma. N Engl J Med 324 (11): 709-15, 1991<br />

10. Hill MJ, Morson BC, Bussey HJ: Aetiology <strong>of</strong> adenoma--carcinoma sequence<br />

in large bowel. Lancet 1 (8058):245-7, 1978<br />

11. Potter JD: Reconciling the epidemiology, physiology, and molecular biology<br />

<strong>of</strong> colon cancer. JAMA 268 (12): 1573-7, 1992 Sep 23-30<br />

12. Winawer, SJ, Fletcher, RH, Miller, L, et al. Colorectal cancer screening: clinical<br />

guidelines and rationale. Gastroenterology 1997; 112:594<br />

13. Winawer, SJ, Zauber,AG, Ho, MN, et al. Prevention <strong>of</strong> colorectal cancer by<br />

colonoscopic polypectomy. The National Polyp Study Work Group. N Engl J Med<br />

1993; 329:1977<br />

14. Burt, RW, DiSario, JA, Cannon-Albright, L. Genetics <strong>of</strong> colon cancer: Impact<br />

<strong>of</strong> inheritance on colon cancer risk. Annu Rev Med 1995; 46:371<br />

15. Spirio, L, Olschwang, S, Groden, J. Alleles <strong>of</strong> the APC gene: An attenuated<br />

form <strong>of</strong> familial polyposis. Cell 1993; 75:951<br />

16. Lynch, HT, Smyrk, TC, Watson, P, et al. Genetics, natural history, tumor<br />

spectrum, and pathology <strong>of</strong> hereditary nonpolyposis colorectal cancer: An updated<br />

review. Gastoenterology 1993; 104:1535<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 71<br />

70<br />

70<br />

Manage per findings<br />

Manage as per<br />

special scenarios<br />

Genetic counseling<br />

17. Atkin, WS, Morson, BC, Cuzick, J. Long-term risk <strong>of</strong> colorectal cancer after<br />

excision <strong>of</strong> rectosigmoid adenomas. N Engl J Med 1992; 326:658<br />

18. Winawer, SJ, Zauber, AG, Gerdes, H, et. al. Risk <strong>of</strong> colorectal caner in the<br />

families <strong>of</strong> patients with adenomatous polyps. National Polyp Study Workgroup.<br />

N Engl J Med 1996; 334:82<br />

19. Ekbom, A, Helmick, C, Zack, M, et. al. Ulcerative colitis and colorectal cancer:<br />

A population-based study. N Engl J Med 1990; 323:1228<br />

20. Brentnall, TA, Haggitt, RC, Rabinovitch, PS, et. al. Risk and natural history<br />

colonic neoplasia in patients with primary sclerosing cholangitis and ulcerative<br />

colitis. Gastroenterology 1996; 110:331<br />

21. D’Haens, GR, Lashner, BA, Hanauer, SB. Pericholongitis and sclerosing<br />

cholangitis are risk factors for dysplasia and cancer in ulcerative colitis. Am J<br />

Gastroenterol 1993; 88:1174<br />

22. Inoue, M, Iwasaki, M, Otani, T, et al. Diabetes mellitus and the risk <strong>of</strong> cancer:<br />

results from a large-scale population-based cohort study in Japan. Arch Intern<br />

Med 2006; 166:1871<br />

23. Ragozzino, M, Melton, LJ 3d, Chu, CP, Palumbo, PJ. Subsequent cancer risk<br />

in the incidence cohort <strong>of</strong> Rochester, Minnesota, residents with diabetes mellitus.<br />

J. Chronic Dis 1982; 35:13<br />

24. La Vecchia, C, Negri, E, Decarli, A, Franceschi, S. Diabetes mellitus and<br />

colorectal cancer risk. Cancer Epidemiol Biomarkers Prev 1997; 6:1007<br />

25. Hu, FB, Mansion, JE, Liu, S, et al. Prospective study <strong>of</strong> adult onset diabetes<br />

mellitus (Type 2) and risk <strong>of</strong> colorectal cancer in women. J Natl Cancer Inst<br />

1999; 91:542<br />

26. Jee, SH, Ohrr, H, Sull, JW, et al. Fasting serum glucose level and cancer risk<br />

in Korean men and women. JAMA 2005; 293;194<br />

27. Yang, YX, Hennessy, S, Lewis, JD. Type 2 diabetes mellitus and the risk <strong>of</strong><br />

colorectal cancer. Clin Gastroenterol Hepatol 2005; 3:587<br />

28. Lagergren, J, Ye, W, Ekbom, A. Intestinal cancer after cholecystectomy: Is bile<br />

involved in carcinogenesis?. Gastroenterology 2001; 121:542<br />

29. Giovannucci, E, Colditz, GA, Stampfer, MJ. A meta-analysis <strong>of</strong> cholecystectomy<br />

and risk <strong>of</strong> colorectal cancer. Gastroenterology 1993: 105:130<br />

30. Reid, FD, Mercer, PM, Harrison, M, Bates, T. Cholecystectomy as a risk<br />

factor for colorectal cancer: A meta-analysis. Scand J Gastroenterol 1996; 31:160<br />

31.Todoroki, I, Friedman, GD, Slattery, ML, et al. Cholecystectomy and the risk


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

<strong>of</strong> colon cancer. Am J Gastroenterol 1999; 94:41<br />

32. Mercer, PM, Reid, FD, Harrison, M, Bates, T. The relationship between<br />

cholecystectomy, unoperated gallstone disease, and colorectal cancer. A necropsy<br />

study. Scand J Gastroenterol 1995; 30:1017<br />

33. Longneck, MP, Orza, MJ, Adams, ME, et al. A meta-analysis <strong>of</strong> alcoholic<br />

beverage consumption in relation to risk <strong>of</strong> colorectal cancer. Cancer Causes<br />

Control 1990; 1:59<br />

34. Gapstur, SM, Potter, JD, Folsom, AR. Alcohol consumption and colon and<br />

rectal cancer in postmenopausal women. Int J Epidemiol 1994; 23:50<br />

35. Thun, MJ, Peto, R, Lopez, AD, et al. Alcohol consumption and mortality among<br />

middle-aged and elderly U.S. adults. N Engl J Med 1997; 337:1705<br />

36. Pedersen, A, Johansen, C, Gronbaek, M. Relations between amount and type<br />

<strong>of</strong> alcohol and colon and rectal cancer in a Danish population based cohort<br />

study. Gut 2003; 52:861<br />

37. Shimizu, N, Nagata, C, Shimizu, H, et al. Height, weight, and alcohol<br />

consumption in relation to the risk <strong>of</strong> colorectal cancer in Japan: a prospective<br />

study. Br J Cancer 2003; 88:1038<br />

38. Ye, W, Romelsjo, A, Augustsson, K, Adami, HO. No excess risk <strong>of</strong> colorectal<br />

cancer among alcoholics followed for up to 25 years. Br J Cancer 2003; 88:1044<br />

39. Cho, E, Smith-Warner, SA, Ritz, J, et al. Alcohol intake and colorectal cancer<br />

a pooled analysis <strong>of</strong> 8 cohort studies. Ann Intern Med 2004; 140:603<br />

40. Martinez, ME, Giovannucci, E, Spiegelman, D,et al. Leisure-time physical<br />

activity, body size, and colon cancer in women. Nurses’ Health Study Research<br />

Group. J Natl Cancer Inst 1997,; 89:948<br />

41. Giovannucci, E, Ascherio, A, Rimm, EB, et al. Physical activity obesity, and<br />

risk for colon cancer and adenoma in men. Ann Intern Med 1995; 122:327<br />

42. Calle, EE, Rodriguez, C, Walker-Thurmond, K, Thun, MJ. Overnight, obesity,<br />

and mortality from cancer in a prospectively studied cohort <strong>of</strong> U.S. adults. N Engl<br />

J Med 2003; 348:1625.<br />

43. Dignam, JJ, Polite, BN, Yothers, G, et al. Body mass index and outcomes<br />

in patients who receive adjuvant chemotherapy for colon cancer. J Natl Cancer<br />

Inst 2006; 98:1647.<br />

44. Chan, AO, Jim, MH, Lam, KF, et al. Smoking and colorectal cancer: A metaanalysis.<br />

JAMA 2008; 300:2765.<br />

45. Botteri, E, Iodice, S, Raimondi, S, et al. Cigarette smoking and adenomatous<br />

polyps: A meta-analysis. JAMA 2008; 300:2765.<br />

46. Botteri, E, Iodice, S, Raimondi, S, et al. Cigarette smoking and adenomatous<br />

polyps: A meta-analysis. Gastroentorology 2008; 134:388.<br />

47. Delhougne, B, Deneux, C, Abs, R, et al. The prevalence <strong>of</strong> colonic polyps<br />

in acromegaly: A colonoscopic and pathological study in 103 patients. J Clin<br />

Endcorinol Metab 1995; 80:3223.<br />

48. Fukuda, I, Hizuka, N, Murakami, Y, Itoh, E. Clinical features and therapeutic<br />

outcomes 0f 65 patients with acromegaly at Tokyo Women’s <strong>Medical</strong> University.<br />

Intern Med 2001; 40:987<br />

49. Chao, A, Thun, MJ, Connel, CJ, et al. Meat consumption and risk <strong>of</strong> colorectal<br />

cancer. JAMA 2005; 293:172.<br />

50. Norat, T, Bingham, S, Ferrari, P, et al. Meat, fish and colorectal cancer risk:<br />

The European Prospective Investigation into cancer and nutrition. J Natl Cancer<br />

Inst 2005;97:906.<br />

51. Willett, WC, Stampfer, MJ, Colditz, GA, et al. Relation <strong>of</strong> meat, fat and fiber<br />

intake to the risk <strong>of</strong> colon cancer in a prospective study among women. N Engl<br />

J Med 1990;323:1664.<br />

52. Sandhu, MS, White, IR, McPherson, K, Systemic review <strong>of</strong> the prospective<br />

cohort studies on meat consumption and colorectal cancer risk: A meta-analytical<br />

approach. Cancer Epidemiol Biomarkers Prev 2001; 10:439.<br />

53. MacLennan, R, Macrae, F, Bain, C, et al. Randomized trial <strong>of</strong> intake <strong>of</strong><br />

fat, fiber and beta carotene to prevent colorectal adenomas. J Natl Cancer Inst<br />

2995;87:1760.<br />

54. Giovannucci, E. Meta-analysis <strong>of</strong> c<strong>of</strong>fee consumption and risk <strong>of</strong> colorectal<br />

cancer. Am J Epidemiol 1998; 147:1043.<br />

55. Michel, KB, Willett, WC, Fuchs, CS, Giovannucci, e. C<strong>of</strong>fee, tea and caffeine<br />

consumption and incidence <strong>of</strong> colon and rectal cancer. J Natl Cancer Inst 2005;<br />

97:282.<br />

56. Baxter, NN, Tepper, JE, Durham, SB, et al. Increased risk <strong>of</strong> rectal cancer after<br />

prostate radiation. A population-based study. Gastroentorology 2005; 128:819.<br />

57. Bini, EJ, Park, J, Francois, F. Use <strong>of</strong> flexible sigmoidoscopy to screen for<br />

colorectal cancer in HIV-infected patients 50 years <strong>of</strong> age and older. Arch Intern<br />

Med 2006; 166:1626.<br />

58. Clifford, GM, Polesel, J, Richenbach, M, et al. Cancer risk in the Swiss HIB<br />

Cohort Study: association with immunodeficiency, smoking, and highly active<br />

antiretroviral therapy. J Natl Cancer Inst 2005; 97:425<br />

59. Hodgson, DC, Gilbert, ES, Dores, GM, et al. Long-term solid cancer risk<br />

among 5-year survivors <strong>of</strong> Hodgkin’s lymphoma. J. Clin Oncol 2007; 25:1489<br />

60. Kim, YI, Mason, JB. Nutritional chemoprevention <strong>of</strong> gastrointestinal cancer:<br />

A critical review. Nutr Rev 1996; 54:259<br />

61. Terry, P, Giovannucci, E, Michels, KB, et al. Fruit, vegetables, dietary fiber,<br />

and risk <strong>of</strong> colorectal cancer. J Natl Cancer Inst 2001; 93:252<br />

62. Slattery, ML, Boucher, KM, Caan, BJ, et al. Eating patterns and risk <strong>of</strong> colon<br />

cancer. Am J Epidemiol 1998; 148:4<br />

63. Negri, E, Franceschi, S, Parpinel, M, La Vecchia, C. Fiber intake and risk <strong>of</strong><br />

colorectal cancer. Cancer Epidemiol Biomarkers Prev 1998; 7:667<br />

64. Wei, EK, Giovannucci, E, Selhub, J, et al. Plasma vitamin B6 and the risk<br />

colorectal cancer and adenoma in women. J Natl Cancer Inst 2005; 97:684<br />

65. Holt, PR, Atillasoy, EO, Gilman, J, et al. Modulation <strong>of</strong> abnormal colonic<br />

epithelial cell proliferation and differentiation by low-fat diary foods : A randomized<br />

controlled trial. JAMA 1998; 280:1074<br />

66. Baron, JA, Beach, M, Mandel, JS, et al. Calcium supplements for the prevention<br />

<strong>of</strong> colorectal adenomas. Calcium Polyp Prevention Study Group. N Engl J Med<br />

1999; 340:101.<br />

67. Bond, JH. Polyp guideline: Diagnosis, treatment and surveillance for patients<br />

with colorectal polyps. Practice Parameters Committee <strong>of</strong> the American College<br />

<strong>of</strong> Gastroentorology. Am J Gastroenterol 2000; 95:3053.<br />

68. Wu, K, Willett, WC, Fuchs, CS, et al. Calcium intake and risk <strong>of</strong> colon cancer<br />

in women and men. J Natl Cancer Inst 2002; 94:437<br />

69. Kim, HS, Newcomb, PA, Ulrich, CM, et al. Vitamin D receptor polymorphism<br />

and the risk <strong>of</strong> colorectal adenomas: Evidence <strong>of</strong> interaction with dietary vitamin<br />

D and calcium. Cancer Epidemiol Biomarkers Prev 2001; 10:869<br />

70. Weingarten, MA, Zalmanovici, A, Yaphe, J. Dietary calcium supplementation<br />

for preventing colorectal cancer and adenomatous polyps. Cochrane Database<br />

Syst Rev 2004; CD003548<br />

71. Cho, E, Smith-Warner, SA, Spiegelman, D, et al. Diary foods, calcium, and<br />

colorectal cancer: a pooled analysis <strong>of</strong> 10 cohort studies. J Natl Cancer Inst<br />

2004:96:1015<br />

72. Wactawski-Wende, J, Kotchen, JM, Anderson, GL, et. al. Calcium plus vitamin<br />

D supplementation and the risk <strong>of</strong> colorectal cancer. N engl J Med 2006; 354:684.<br />

73. Larsson, SC, Bergkvist, L, Wolk, A. Magnesium intake in relation to risk <strong>of</strong><br />

colorectal cancer in women. JAMA 2005; 293:86<br />

74. Arber, N, Eagle CJ, Spicak J, et al.: Calecoxib for the prevention <strong>of</strong> colorectal<br />

adenomatous polyps. N Engl J Med 355 (9): 885-95, 2006<br />

75. Solomon, SD, Pfeffer MA, McMurray JJ, et al.: Effect <strong>of</strong> celecoxib on<br />

cardiovascular events and blood pressure in two trials for the prevention <strong>of</strong><br />

colorectal adenomas. Circulation 114(10): 1028-35, 2006<br />

72 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


76. Colditz, GA, Cannusicio, CC, Frazier, AL. Physical activity and reduced risk<br />

<strong>of</strong> colon cancer: Implications for prevention. Cancer Causes Control 1997; 8:649<br />

77. Mao, Y, <strong>Pan</strong>, S, Wen, SW, Johnson, KC. Physical inactivity, energy intake,<br />

obesity and the risk <strong>of</strong> rectal cancer in Canada. Int J Cancer 2003; 105:831<br />

78. Colbert, LH, Hartman, TJ, Malila, N, et al. Physical activity in relation to<br />

cancer <strong>of</strong> the colon and rectum in a cohort <strong>of</strong> male smokers. Cancer Epidemiol<br />

Biomakers Prev 2001; 10:265<br />

79. Meyskens, FL, Gerner, EW, Peolot, D, et al. Marked efficacy <strong>of</strong><br />

diflouromethylornithine plus sulindac in reducing recurrent adenomas in patients<br />

with prior adenomas. Results <strong>of</strong> a randomized, placebo--controlled, double blind<br />

Phase III trial (abstract). In: Proceedings <strong>of</strong> the 99th Annual Meeting <strong>of</strong> the<br />

American <strong>Association</strong> for Cancer Research 2008; Apr 12-16; San Diego, CA.<br />

Philadelphia (PA): AACR; 2008. Abstract LB-142<br />

80. Grodstein, F, Martinez, ME, Platz, EA, et al. Postmenopausal hormone use<br />

and the risk for colorectal cancer and adenoma. Ann Intern Med 1998; 128:705<br />

81. Calle, EE, Miraclemcmahill, HL, Thun, MJ, Health, CW. Estrogen replacement<br />

therapy and risk <strong>of</strong> fatal colon cancer in a prospective cohort <strong>of</strong> postmenopausal<br />

women. J Natl Cancer Inst. 1995; 87:51<br />

82. Grodstein, F, Newcomb, PA, Stampfer, MJ. Postmenopausal hormone<br />

therapy and the risk <strong>of</strong> colorectal cancer: A review and meta-analysis. Am J<br />

Med 1999;106:574.<br />

83. Slattery, ML, Potter, JD, Curtin, K, et al. Estrogens reduce and withdrawal <strong>of</strong><br />

estrogens increase risk <strong>of</strong> microsatellite instability-positive colon cancer. Cancer<br />

Res 2001;61: 126.<br />

84. Chlebowski, RT, Wactawski-Wende, J, Ritenbaugh, C, et al. Estrogen plus<br />

progestin and colorectal cancer in postmenopausal women. N Engl J Med 2004;<br />

350: 9941.<br />

85. Anderson, GL, Limacher, M, Assaf, AR, et al. Effects <strong>of</strong> conjugated equine<br />

estrogen in postmenopausal women with hysterectomy: the Women’s Health<br />

Initiative randomized controlled trial. JAMA 2004; 291: 1701.<br />

86. Woodson, K, Lanza, E, Tangrea, JA, et al. Hormone replacement therapy and<br />

colorectal adenoma recurrence among women in the polyp prevention trial. J Natl<br />

Cancer Inst 2001; 93: 1799.<br />

87. Sacks, FM, Pfeffer, MA, Moye, LA, et al. The effect <strong>of</strong> pravastatin on coronary<br />

events after myocardial infarction in patients with average cholesterol levels.<br />

Cholesterol and Recurrent Events Trial investigator. N Engl J Med 1996; 335:1001.<br />

88. Pedersen, TR, Berg, K, Cook, TJ, et al. Safety and tolerability <strong>of</strong> cholesterol<br />

lowering with simvastatin during 5 years in the Scandinavian Simvastatin Survival<br />

Study. Arch lnter Med 1996; 156:2085<br />

89.Poynter, JN, Gruber, SB, Higgins, PD, et al. Statins and the risk <strong>of</strong> colorectal<br />

cancer. N Eng J Med 2005; 352:2184<br />

90. Bjelakovic, G, Nagorni, A, Nikolova, D, et al. Meta-analysis: antioxidant<br />

supplement for primary and secondary prevention <strong>of</strong> colorectal adenoma. Aliment<br />

Pharmacol Ther2006; 24:281.<br />

91.Geelen, A, Schouten, JM, Kamphuis, C, et al. Fish consumption, n-3 fatty<br />

acid, and colorectal cancer: a meta-analysis <strong>of</strong> prospective cohort studies. Am J<br />

Epidemiol 2007; 166:116.<br />

92. Ngo, SN, Wiliams, DB, Cobiac, L, Head, RJ. Does garlic reduce risk <strong>of</strong><br />

colorectal cancer? A systematic review. J Nutr 2007; 137:2264.<br />

93.World Cancer Research Fund/American Institute <strong>of</strong> Cancer Research. Food,<br />

Nutrition, Physical Activity and the Prevention <strong>of</strong> Cancer: a Global Perspective.<br />

Washington DC: AICR, 2007.<br />

94.Imperiale, TF, Wagner, DR, Lin, CY, et al. Results <strong>of</strong> screening colonoscopy<br />

among persons 40 to 49 years <strong>of</strong> age. N Eng J Med 2002; 346:1781<br />

95.Increased use <strong>of</strong> colorectal cancer tests—United States, 2002 and 2004. MMVVR<br />

Morb Mortal Wkly Rep 2006;55:308<br />

96. Winawer, SJ, Fletcher, RH, Miler, L, et al. Colorectal cancer screening: clinical<br />

guidelines and rationale. Gastroenterology 1997; 112: 5974.<br />

97.Hawkin, NJ, Ward, RL. Sporadic colorectal cancers with microsatellite<br />

instability and their possible origin in hyperplasic Polyps serrated adenomas. J<br />

Natl Cancer Inst 2001; 93 1307.<br />

98. Winawer, SJ, Zauber, AG, Ho, MN, et al. Prevention <strong>of</strong> colorectal cancer by<br />

colonoscopic polypectomy. The National Polyp Study Work Group. N Eghl Med<br />

1993;329:1977.<br />

99. Winawer, SJ, Fletcher, RH, Miler, L, et al. Colorectal cancer screening: clinical<br />

guidelines and rationale. Gastroenterology 1997; 112: 594.<br />

100. Umar, A, Boland, CR, Terdiman, JP, et al. Revised Bethesada Guidelines for<br />

hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite<br />

instability. J Natl Cancer Inst 2004; 96; 261<br />

101.Winawer, S, Fletcher, R, Rex, D, et al. colorectal cancer screening and<br />

surveillance: clinical guidelines and rationale-Update based on new evidence.<br />

Gastroenterology 2003; 124: 544.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 73


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

PROSTATE CANCER SCREENING<br />

Dr. Alaa Kandil<br />

Alexandria School <strong>of</strong> Medicine, Alexandria, Egypt<br />

Corresponding Author: Dr. Alaa Kandil<br />

Pr<strong>of</strong>essor <strong>of</strong> Clinical <strong>Oncology</strong>, Alexandria School <strong>of</strong> Medicine<br />

Alexandria, Egypt<br />

E-mail: alaakandil@hotmail.com<br />

Abstract<br />

Background: Prostate cancer is a very common malignancy affecting males<br />

worldwide. There is a need to understand its risk factors, early detection and<br />

prevention approach to this disease.<br />

Methods: Prostate cancer risk factors, early detection were reviewed from the<br />

literature and the available guidelines for screening were reviewed and summarized.<br />

Results: Prostate cancer early detection can be done by performing digital rectal<br />

examination and obtaining prostate specific antigen (PSA). Although generally<br />

the screening is done after age 50, in some higher risk group, it should be done<br />

at younger age. However, this has to be done after discussion with the patients<br />

each individual situation. General public screening is controversial and there is<br />

no prove <strong>of</strong> impact on survival.<br />

Conclusion: Screening for prostate cancer should be individualized and patients<br />

should be counseled about it prior to performing the test.<br />

Prostate cancer is the most common cancer diagnosed in North American men,<br />

excluding skin cancers. It is estimated that in 2008, approximately 186,320 new<br />

cases and 28,660 prostate cancer-related deaths will occur in the United States.<br />

Prostate cancer is now the second leading cause <strong>of</strong> cancer death in men, exceeded<br />

only by lung cancer. It accounts for 25% <strong>of</strong> all male cancers and 10% <strong>of</strong> male<br />

cancer-related deaths.(1)<br />

Regional differences have been observed in prostate cancer incidence and mortality<br />

rates and in rates <strong>of</strong> radical prostatectomy. The increased incidence until 1989<br />

was most likely the result <strong>of</strong> increased tumor detection due to increasing rates<br />

<strong>of</strong> transurethral prostatectomy(2, 3) Subsequent increases were most likely the<br />

result <strong>of</strong> widespread use <strong>of</strong> PSA testing for early detection and screening.(4, 5)<br />

Risk Factors<br />

Prostate cancer is uncommonly seen in men younger than 50 years; the incidence<br />

rises rapidly with each decade thereafter. The age-adjusted incidence is higher<br />

in African American males (258.3 per 100,000) than in white males (163.4 per<br />

100,000)(6)<br />

African American males have a higher mortality from prostate cancer, even after<br />

attempts to adjust for access-to-care factors.(7)<br />

Men with a family history <strong>of</strong> prostate cancer are at an increased risk <strong>of</strong> the disease<br />

compared with men without this history.(8, 9)<br />

Other potential risk factors besides age, race, and family history <strong>of</strong> prostate cancer<br />

include alcohol consumption, vitamin or mineral interactions, and other dietary<br />

habits.(10 – 14)<br />

There is an association between primary tumor volume and local extent <strong>of</strong> disease,<br />

progression, and survival.(15)<br />

A review <strong>of</strong> a large number <strong>of</strong> prostate cancers in radical prostatectomy, cystectomy,<br />

and autopsy specimens showed that capsular penetration, seminal vesicle invasion,<br />

and lymph node metastases were usually found only with tumors larger than<br />

1.4 cc.(16) Furthermore, the semiquantitative histopathologic grading scheme<br />

proposed by Gleason is reasonably reproducible among pathologists and correlates<br />

with the incidence <strong>of</strong> nodal metastases and with patient survival in a number <strong>of</strong><br />

reported studies.(17)<br />

Cancer statistics from the American Cancer Society and the National Cancer<br />

Institute indicated in 2004 that the proportion <strong>of</strong> disease diagnosed at a locoregional<br />

stage and at a distant stage is 91% and 5% for whites, compared with 89% and<br />

7% for African Americans, respectively.(18) Stage distribution <strong>of</strong> prostate cancer<br />

is affected substantially by the intensity <strong>of</strong> early detection efforts.<br />

With the proliferation <strong>of</strong> PSA for early detection, reviews <strong>of</strong> large numbers <strong>of</strong><br />

asymptomatic men with prostate cancer found that most have organ-confined<br />

disease. One study found that 63% <strong>of</strong> cancers detected in men undergoing their<br />

first screening PSA were pathologically organ-confined cancers; the percentage<br />

increased to 71% if cancer was detected on a subsequent examination. (19)<br />

Screening<br />

Before the 1990s, the digital rectal examination (DRE) was the test traditionally<br />

used for prostate cancer screening. Two other procedures are also available:<br />

transrectal ultrasound (TRUS) imaging and serum prostate-specific antigen (PSA)<br />

concentrations.(20)<br />

Digital Rectal Exam (DRE)<br />

Although DRE has been used for many years, careful evaluation <strong>of</strong> this modality<br />

has yet to take place. Several observational studies have examined process measures<br />

such as sensitivity and case-survival data, but without appropriate controls and<br />

with no adjustment for lead-time and length biases.(21, 22)<br />

Since PSA assays became widely available in the late 1980s, DRE alone is rarely<br />

discussed as a screening modality. A number <strong>of</strong> studies have found that DRE has<br />

a poor predictive value for prostate cancer if PSA is at very low levels.<br />

In the European Study on Screening for Prostate Cancer, it was found that if DRE<br />

is used only for a PSA higher than 1.5 ng/mL (thus, no DRE is performed with<br />

PSA < 1.5 ng/mL), 29% <strong>of</strong> all biopsies would be eliminated while maintaining<br />

a 95% prostate cancer detection sensitivity. By applying DRE only for patients<br />

with a PSA higher than 2.0 ng/m, the biopsy rate would decrease by 36% while<br />

sensitivity would drop to only 92%.(23)<br />

A previous report from this same institution found DRE to have poor performance<br />

characteristics. Among 10,523 men randomly assigned to screening, it was reported<br />

that the overall prostate cancer detection rate using PSA, DRE, and TRUS was<br />

4.5% compared with only 2.5% if DRE alone had been used. Among men with a<br />

PSA lower than 3.0 ng/mL, the PPV <strong>of</strong> DRE was only 4% to 11%. (24)<br />

Rectal examination is inexpensive, relatively noninvasive, and nonmorbid and can<br />

be taught to nonpr<strong>of</strong>essional health workers; however, its effectiveness depends<br />

on the skill and experience <strong>of</strong> the examiner. The possible contribution <strong>of</strong> routine<br />

annual screening by rectal examination to reducing prostate cancer mortality<br />

remains to be determined.<br />

Transrectal Ultrasound and Other Imaging Tests<br />

Imaging procedures have been suggested as possible screening modalities for prostate<br />

cancer. Prostatic imaging is possible by ultrasound, computed tomography, and<br />

magnetic resonance imaging. Each modality has relative merits and disadvantages for<br />

distinguishing different features <strong>of</strong> prostate cancer. Ultrasound has received the most<br />

attention, having been examined by several investigators in observational settings.(25)<br />

74 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


Sensitivity ranged from 71% to 92% for prostatic carcinoma and 60% to 85%<br />

for subclinical disease. Specificity values ranged from 49% to 79%, and positive<br />

predictive values in the 30% range have been reported. The sensitivity and positive<br />

predictive value for ultrasound as a single test may be better than for rectal<br />

examination. The rate <strong>of</strong> cancer is extremely low among ultrasound-positive<br />

patients in whom rectal and PSA examinations are normal.(26)<br />

Contemporary prostate biopsy relies on spring-loaded biopsy devices that are either<br />

digitally guided or guided via ultrasound. TRUS guidance is the most frequently<br />

used method <strong>of</strong> directing prostate needle biopsy because there is some suggestion<br />

that the yield <strong>of</strong> biopsy is improved with such guidance.(27)<br />

With the virtually simultaneous clinical acceptance <strong>of</strong> TRUS, spring-loaded biopsy<br />

devices, and the proliferation <strong>of</strong> PSA screening in the late 1980s, the number <strong>of</strong><br />

prostate cores obtained from patients with either an abnormal DRE or PSA was<br />

most commonly six, using a sextant method <strong>of</strong> sampling the prostate.(28)<br />

There is evidence that the predictable increase in cancer detection rates that would<br />

be expected by increasing the number <strong>of</strong> biopsy cores beyond six does occur;<br />

e.g., biopsies with 12 or 15 cores would increase the proportion <strong>of</strong> biopsied men<br />

having cancer detected by 30% to 35%.(29, 30)<br />

The extent to which such increased detection will reduce morbidity and mortality<br />

from the disease or increase the fraction <strong>of</strong> men treated unnecessarily is unknown.<br />

Prostate-Specific Antigen (PSA)<br />

The PSA test has been examined in several observational settings for initial<br />

diagnosis <strong>of</strong> disease, as a tool to monitor for recurrence after initial therapy, and<br />

for prognosis <strong>of</strong> outcomes after therapy. There is no PSA value below which a<br />

man can be assured that he has no risk <strong>of</strong> prostate cancer. Parameter estimates for<br />

this test include sensitivity in the range <strong>of</strong> 70%.(31)<br />

The potential value <strong>of</strong> the test appears to be in its simplicity, objectivity,<br />

reproducibility, relative lack <strong>of</strong> invasiveness, and relatively low cost. PSA has<br />

increased the detection rate <strong>of</strong> early-stage cancers, many <strong>of</strong> which may be curable<br />

by local-modality therapies.(32-35)<br />

Experience with repeat PSA screening suggests that tumors detected on follow-up<br />

examinations are <strong>of</strong> lower clinical stage and grade.(36)<br />

Although a cut<strong>of</strong>f value <strong>of</strong> 4.0 ng/mL is frequently used to prompt prostate biopsy,<br />

screening studies have demonstrated that lowering the PSA cut<strong>of</strong>f will substantially<br />

increase the number <strong>of</strong> cancers detected, particularly in African Americans.(37)<br />

An initial PSA lower than 2.5 ng/mL is associated with a very low risk <strong>of</strong> cancer<br />

detection within a 4-year follow-up.(36-38)<br />

The Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial (PLCO) is<br />

a multicenter, randomized, two-armed trial designed to evaluate the effect <strong>of</strong><br />

screening for prostate, lung, colorectal, and ovarian cancer on disease-specific<br />

mortality. Enrollment began in November 1993 and concluded in June 2001.<br />

Participants were randomly assigned to the screening or control arm. A total <strong>of</strong><br />

76,705 men were enrolled, 38,250 <strong>of</strong> whom were assigned to the screening arm. Of<br />

these, 34,244 men underwent an initial PSA and/or DRE screening examination.<br />

Compliance rates for PSA and DRE were roughly equivalent at 89%. More than<br />

99% <strong>of</strong> men who underwent screening with either PSA or DRE received both<br />

screening tests.(39)<br />

Table 1: Summary <strong>of</strong> Prostate, Lung, Colon and Ovarian (PLCO) Screening<br />

Trial Results<br />

Screening Test PSA DRE PSA or DRE<br />

Administered N0 (%) N0 (%) N0 (%)<br />

Number <strong>of</strong> Men<br />

Receiving Test<br />

34, 233 34, 115 34, 224<br />

Positive Test (%): DRE<br />

Suspicious for Cancer;<br />

PSA >4 ng/mL<br />

2, 717 (7.9) 2, 482 (7.3) 4, 801 (14.1)<br />

Biopsies (% <strong>of</strong><br />

positives)<br />

1, 112 (40.9) 639 (25.7) 1, 510 (31.5)<br />

Prostate Cancer (% <strong>of</strong><br />

biopsies)<br />

489 (44.0) 219 (34.3) 556 (36.8)<br />

Prostate Cancer (% <strong>of</strong><br />

positives)<br />

489 (18.0) 219 (8.8) 556 (11.6)<br />

PSA: Prostate specific Antigen. DRE: Digital Rectal Examination<br />

Various approaches aimed at improving the performance <strong>of</strong> PSA in early cancer<br />

detection have been tested. None are clearly more accurate than total serum PSA<br />

levels, but these approaches are listed below.<br />

• Complexed PSA and percent-free PSA<br />

• Third-generation PSA<br />

• PSA density<br />

• PSA density <strong>of</strong> the transition zone<br />

• Age-adjusted PSA<br />

• PSA velocity<br />

Frequency <strong>of</strong> screening<br />

The optimal frequency and age range for PSA (and DRE) testing are unknown<br />

(40, 41). Cancer detection rates have been reported to be similar for intervals <strong>of</strong><br />

1 to 4 years. (42)<br />

Table 2: American Cancer Society Recommendation<br />

Population Age Performing PSA,<br />

DRE<br />

Average risk ≥ 50<br />

Moderate high risk<br />

≥ 45<br />

(one first degree<br />

relative with prostate<br />

cancer


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

5. Jacobsen SJ, Katusic SK, Bergstralh EJ, et al.: Incidence <strong>of</strong> prostate cancer<br />

diagnosis in the eras before and after serum prostate-specific antigen testing.<br />

JAMA 274 (18): 1445-9, 1995.<br />

6. Ries LAG, Harkins D, Krapcho M, et al.: SEER Cancer Statistics Review,<br />

1975-2003. Bethesda, Md: National Cancer Institute, 2006.<br />

7. Robbins AS, Whittemore AS, Van Den Eeden SK: Race, prostate cancer survival,<br />

and membership in a large health maintenance organization. J Natl Cancer Inst<br />

90 (13): 986-90, 1998.<br />

8. Steinberg GD, Carter BS, Beaty TH, et al.: Family history and the risk <strong>of</strong><br />

prostate cancer. Prostate 17 (4): 337-47, 1990.<br />

9. Matikainen MP, Schleutker J, Mörsky P, et al.: Detection <strong>of</strong> subclinical cancers<br />

by prostate-specific antigen screening in asymptomatic men from high-risk prostate<br />

cancer families. Clin Cancer Res 5 (6): 1275-9, 1999.<br />

10. Hayes RB, Brown LM, Schoenberg JB, et al.: Alcohol use and prostate cancer<br />

risk in US blacks and whites. Am J Epidemiol 143 (7): 692-7, 1996.<br />

11. Platz EA, Leitzmann MF, Rimm EB, et al.: Alcohol intake, drinking patterns,<br />

and risk <strong>of</strong> prostate cancer in a large prospective cohort study. Am J Epidemiol<br />

159 (5): 444-53, 2004.<br />

12. Eichholzer M, Stähelin HB, Gey KF, et al.: Prediction <strong>of</strong> male cancer mortality<br />

by plasma levels <strong>of</strong> interacting vitamins: 17-year follow-up <strong>of</strong> the prospective<br />

Basel study. Int J Cancer 66 (2): 145-50, 1996.<br />

13. Gann PH, Hennekens CH, Sacks FM, et al.: Prospective study <strong>of</strong> plasma<br />

fatty acids and risk <strong>of</strong> prostate cancer. J Natl Cancer Inst 86 (4): 281-6, 1994.<br />

14. Morton MS, Griffiths K, Blacklock N: The preventive role <strong>of</strong> diet in prostatic<br />

disease. Br J Urol 77 (4): 481-93, 1996 .<br />

15. Freedland SJ, Humphreys EB, Mangold LA, et al.: Risk <strong>of</strong> prostate cancerspecific<br />

mortality following biochemical recurrence after radical prostatectomy.<br />

JAMA 294 (4): 433-9, 2005.<br />

16. McNeal JE, Bostwick DG, Kindrachuk RA, et al.: Patterns <strong>of</strong> progression in<br />

prostate cancer. Lancet 1 (8472): 60-3, 1986.<br />

17. Resnick MI: Background for screening--epidemiology and cost effectiveness.<br />

Prog Clin Biol Res 269: 111-22, 1988.<br />

18. Ries LAG, Eisner MP, Kosary CL, et al., eds.: SEER Cancer Statistics Review,<br />

1975-2002. Bethesda, Md: National Cancer Institute, 2005.<br />

19. Catalona WJ, Smith DS, Ratliff TL, et al.: Detection <strong>of</strong> organ-confined prostate<br />

cancer is increased through prostate-specific antigen-based screening. JAMA<br />

270 (8): 948-54, 1993.<br />

20. Scardino PT: Early detection <strong>of</strong> prostate cancer. Urol Clin North Am 16 (4):<br />

635-55, 1989.<br />

21. Gilbertsen VA: Cancer <strong>of</strong> the prostate gland. Results <strong>of</strong> early diagnosis and<br />

therapy undertaken for cure <strong>of</strong> the disease. JAMA 215 (1): 81-4, 1971<br />

22. Jenson CB, Shahon DB, Wangensteen OH: Evaluation <strong>of</strong> annual examinations<br />

in the detection <strong>of</strong> cancer: special reference to cancer <strong>of</strong> the gastrointestinal tract,<br />

prostate, breast, and female generative tract. JAMA 174(14): 1783-1788, 1960.<br />

23. Beemsterboer PM, Kranse R, de Koning HJ, et al.: Changing role <strong>of</strong> 3 screening<br />

modalities in the European randomized study <strong>of</strong> screening for prostate cancer<br />

(Rotterdam). Int J Cancer 84 (4): 437-41, 1999.<br />

24. Schröder FH, van der Maas P, Beemsterboer P, et al.: Evaluation <strong>of</strong> the digital<br />

rectal examination as a screening test for prostate cancer. Rotterdam section <strong>of</strong><br />

the European Randomized Study <strong>of</strong> Screening for Prostate Cancer. J Natl Cancer<br />

Inst 90 (23): 1817-23, 1998.<br />

25. Waterhouse RL, Resnick MI: The use <strong>of</strong> transrectal prostatic ultrasonography<br />

in the evaluation <strong>of</strong> patients with prostatic carcinoma. J Urol 141 (2): 233-9, 1989.<br />

26. Cooner WH, Mosley BR, Rutherford CL Jr, et al.: Clinical application <strong>of</strong><br />

transrectal ultrasonography and prostate specific antigen in the search for prostate<br />

cancer. J Urol 139 (4): 758-61, 1988.<br />

27. Renfer LG, Schow D, Thompson IM, et al.: Is ultrasound guidance necessary<br />

for transrectal prostate biopsy? J Urol 154 (4): 1390-1, 1995.<br />

28. Hodge KK, McNeal JE, Stamey TA: Ultrasound guided transrectal core biopsies<br />

<strong>of</strong> the palpably abnormal prostate. J Urol 142 (1): 66-70, 1989.<br />

29. Levine MA, Ittman M, Melamed J, et al.: Two consecutive sets <strong>of</strong> transrectal<br />

ultrasound guided sextant biopsies <strong>of</strong> the prostate for the detection <strong>of</strong> prostate<br />

cancer. J Urol 159 (2): 471-5; discussion 475-6, 1998.<br />

30. Eskew LA, Bare RL, McCullough DL: Systematic 5 region prostate biopsy is<br />

superior to sextant method for diagnosing carcinoma <strong>of</strong> the prostate. J Urol 157<br />

(1): 199-202; discussion 202-3, 1997.<br />

31. Partin AW, Oesterling JE: The clinical usefulness <strong>of</strong> prostate specific antigen:<br />

update 1994. J Urol 152 (5 Pt 1): 1358-68, 1994.<br />

32. Catalona WJ, Smith DS, Ratliff TL, et al.: Detection <strong>of</strong> organ-confined prostate<br />

cancer is increased through prostate-specific antigen-based screening. JAMA<br />

270 (8): 948-54, 1993.<br />

33. Babaian RJ, Mettlin C, Kane R, et al.: The relationship <strong>of</strong> prostate-specific<br />

antigen to digital rectal examination and transrectal ultrasonography. Findings<br />

<strong>of</strong> the American Cancer Society National Prostate Cancer Detection Project.<br />

Cancer 69 (5): 1195-200, 1992 .<br />

34. Brawer MK, Chetner MP, Beatie J, et al.: Screening for prostatic carcinoma<br />

with prostate specific antigen. J Urol 147 (3 Pt 2): 841-5, 1992.<br />

35. Mettlin C, Murphy GP, Lee F, et al.: Characteristics <strong>of</strong> prostate cancers<br />

detected in a multimodality early detection program. The Investigators <strong>of</strong> the<br />

American Cancer Society-National Prostate Cancer Detection Project. Cancer<br />

72 (5): 1701-8, 1993.<br />

36. Smith DS, Catalona WJ, Herschman JD: Longitudinal screening for prostate<br />

cancer with prostate-specific antigen. JAMA 276 (16): 1309-15, 1996 Oct 23-30.<br />

37. Smith DS, Carvalhal GF, Mager DE, et al.: Use <strong>of</strong> lower prostate specific<br />

antigen cut<strong>of</strong>fs for prostate cancer screening in black and white men. J Urol 160<br />

(5): 1734-8, 1998.<br />

38. Moul JW, Connelly RR, Mooneyhan RM, et al.: Racial differences in tumor<br />

volume and prostate specific antigen among radical prostatectomy patients. J<br />

Urol 162 (2): 394-7, 1999.<br />

39. Andriole GL, Levin DL, Crawford ED, et al.: Prostate cancer screening in<br />

the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial:<br />

findings from the initial screening round <strong>of</strong> a randomized trial. J Natl Cancer<br />

Inst 97 (6): 433-8, 2005.<br />

40. Etzioni R, Cha R, Cowen ME: Serial prostate specific antigen screening for<br />

prostate cancer: a computer model evaluates competing strategies. J Urol 162<br />

(3 Pt 1): 741-8, 1999.<br />

41. Ross KS, Carter HB, Pearson JD, et al.: Comparative efficiency <strong>of</strong> prostatespecific<br />

antigen screening strategies for prostate cancer detection. JAMA 284<br />

(11): 1399-405, 2000.<br />

42. van der Cruijsen-Koeter IW, Roobol MJ, Wildhagen MF, et al.: Tumor<br />

characteristics and prognostic factors in two subsequent screening rounds with<br />

four-year interval within prostate cancer screening trial, ERSPC Rotterdam.<br />

Urology 68 (3): 615-20, 2006.<br />

76 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


HEPATOCELLULAR CARCINOMA<br />

Dr. Ayman Abdo, MD, FRCPC<br />

King Saud University, Riyadh<br />

Corresponding Author: Dr. Ayman Abdo, MD, AmBIM, FRCPC<br />

Consultant, Gastroentorology Division, College <strong>of</strong> Medicine<br />

Riyadh, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

Disease Epidemiology<br />

Hepatocellular carcinoma (HCC) is the most common primary malignancy <strong>of</strong><br />

the liver. It represents the fifth most common cancer and the third most common<br />

cause <strong>of</strong> cancer death worldwide(4). It has a variable geographical distribution<br />

around the world. The incidence in developing countries is two to three times<br />

higher than in Western countries. For example, in Eastern Asia and Middle Africa<br />

the age-adjusted incidence rate (AAIR) ranges from 20-28 cases per 10,000 in<br />

men while it is about 1-3 per 100,000 in Northern Europe, Australia and North<br />

America. In the United States the incidence <strong>of</strong> HCC has increased from 1.4 per<br />

100,000 population at the period from 1976-1980 to 2.4 per 100,000 population<br />

for the period from 1991-1995.<br />

In Saudi <strong>Arab</strong>ia, according to the National Cancer Registry, liver cancer accounts<br />

for 6.1% <strong>of</strong> all newly diagnosed cancers according to the most recent cancer<br />

registry covering the years 1999-2000. HCC was the second most common cancer<br />

affecting Saudi males and the eighth most common cancer affecting females with<br />

an overall age standardized rate is 4.5/100,000 population. Male to female ratio is<br />

279:100. Of all liver cancers in Saudi <strong>Arab</strong>ia, hepatocellular carcinoma accounts<br />

for 87.6% in Saudi <strong>Arab</strong>ia. The median age at diagnosis is 65 years for males<br />

and 60 years for females.<br />

This incidence <strong>of</strong> HCC in Saudi <strong>Arab</strong>ia is not surprising given the high prevalence<br />

<strong>of</strong> the two major risk factors, namely hepatitis B and hepatitis C infections. In the<br />

large epidemiologic study by Al Faleh et al, 7% <strong>of</strong> Saudi children were found to be<br />

positive for HBsAg. Not until universal vaccination was applied in Saudi <strong>Arab</strong>ia<br />

did this prevalence rate decrease to less than 0.3%. Since the initial epidemiologic<br />

studies showing high prevalence <strong>of</strong> hepatitis B were done on children who are<br />

now adults and with an estimation that about 20% <strong>of</strong> these patients will probably<br />

develop cirrhosis with an annual risk <strong>of</strong> 1-4% for HCC, the incidence <strong>of</strong> HCC is<br />

expected to increase dramatically in the Kingdome in the next 30 years. Hepatitis C<br />

is also common in Saudi <strong>Arab</strong>ia with a prevalence rate <strong>of</strong> 1-3% <strong>of</strong> the population,<br />

which further increases the risk <strong>of</strong> HCC. More recently, the incidence <strong>of</strong> hepatitis<br />

C seems to have decreased to about 1.1%.<br />

Risk Factors<br />

The development <strong>of</strong> cirrhosis is the major risk factor for the development <strong>of</strong> HCC<br />

regardless <strong>of</strong> the cause. The annual incidence <strong>of</strong> HCC in patients with compensated<br />

cirrhosis is about 3%.<br />

Hepatitis B is considered the strongest epidemiologic factor associated with HCC<br />

in the majority <strong>of</strong> countries but more importantly in Asia and Africa. The carrier<br />

state <strong>of</strong> hepatitis B early in life carries a lifetime relative risk <strong>of</strong> developing HCC <strong>of</strong><br />

over 100, while the annual incidence <strong>of</strong> HCC in hepatitis B patients with cirrhosis<br />

exceeds 2%. Hepatitis C is considered the most important risk factor for HCC in<br />

Western countries and Japan. Almost all HCC in patients with hepatitis C occurs<br />

in patients who have developed cirrhosis in which the yearly incidence varies<br />

between 3 to 8 %. Other less likely risk factors include Aflatoxin B1 derived from<br />

Aspergillus flavus and Aspergillous parasiticus which is an important risk factor<br />

for HCC in parts <strong>of</strong> Africa and Asia, hereditary hemochromatosis and Wilson's<br />

disease. Because <strong>of</strong> the significant increase in the prevalence <strong>of</strong> diabetes and<br />

obesity, it is estimated that non alcoholic fatty liver disease may be an important<br />

risk factor for development <strong>of</strong> HCC in the future.<br />

Presenting Signs and Symptoms<br />

The classic features <strong>of</strong> HCC include right upper quadrant pain and weight loss.<br />

Weakness, abdominal swelling, non specific gastrointestinal symptoms, and<br />

jaundice are other presenting features. Special clinical scenarios should raise the<br />

suspicion <strong>of</strong> HCC. This includes acute deterioration <strong>of</strong> liver function in a patient<br />

with stable cirrhosis, new onset ascites, and acute intra-abdominal bleeding.<br />

Physical findings vary according to the stage <strong>of</strong> the disease. If the tumor is small,<br />

no signs may be found except those related to cirrhosis. In more advanced disease,<br />

hepatomegaly is common with a possibility <strong>of</strong> feeling a mass or a hard irregular<br />

liver surface which may be tender. A bruit may be heard on the liver. Ascites is<br />

<strong>of</strong>ten found, most commonly as a result <strong>of</strong> the underlying cirrhosis leading to<br />

portal hypertension but rarely due to tumor invasion <strong>of</strong> the peritoneum. Muscle<br />

wasting is common and is usually progressive.<br />

Screening Methods<br />

Abdominal Ultrasound (US)<br />

While US has the advantage <strong>of</strong> being safe, commonly available, and cost -effective,<br />

its main disadvantage is its low specificity and its operator dependent nature. Newly<br />

discovered focal liver masses in patients with liver cirrhosis has a high likelihood<br />

<strong>of</strong> being HCC. Abdominal US is associated with a sensitivity and specificity <strong>of</strong><br />

20-51% and 92-96%, respectively for detecting lesions consistent with HCC in<br />

patients with cirrhosis. Detection rates for lesions between 2 and 3 cm and 1 and 2<br />

cm with US are estimated to be as low as 20 and 13%, respectively. In spite <strong>of</strong> the<br />

limitations <strong>of</strong> US in diagnosing HCC, due to its low cost, safety, and availability,<br />

it is still considered the best first test to be performed when HCC is expected.<br />

Triphasic CT scan <strong>of</strong> the liver as well as MRI are the diagnostic tests <strong>of</strong> choice in<br />

patients with HCC. Some studies are utilizing these tests in screening purposes<br />

as well especially in high risk patients but high cost and radiation, in case <strong>of</strong> CT,<br />

are significant limiting factors.<br />

Serum Alfa fetoprotein (AFP)<br />

AFP is an alfa 1 globulin that is normally present in high concentrations in fetal<br />

serum but in only minute concentrations in adults. Reported sensitivities are<br />

around 39-65%, specificities around 76-94%, but a poor positive predictive value<br />

<strong>of</strong> 9-50%. In a recent systematic review it was confirmed that AFP has a poor<br />

diagnostic ability to detecting HCC at any level <strong>of</strong> pretest risk. However, as a<br />

confirmatory test in patients with a mass on imaging studies, AFP determination<br />

remains clinically useful. AFP elevation lower than 500 ng/mL may be seen in<br />

patients with active necroinflammatory changes in the liver secondary to active<br />

hepatitis. A progressively rising AFP level even at low concentrations is highly<br />

suggestive <strong>of</strong> HCC. Elevations above 1000 ng/mL have a high specificity rate.<br />

Other tumor markers are under investigation but none is ready for clinical use yet.<br />

These include Des-y-carboxy prothrombin and alfa-l-fucosidase.<br />

Summary <strong>of</strong> Management<br />

Different treatment modalities are available for patients with HCC. The decision<br />

on the best treatment modality should be based on the following factors:<br />

• The number <strong>of</strong> lesions<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 77


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

• The size <strong>of</strong> lesions<br />

• The status <strong>of</strong> the underlying liver<br />

• The status <strong>of</strong> the portal vein<br />

• The patient’s performance status<br />

• The local expertise<br />

• The patient preferences<br />

Current available modalities include: liver transplantation, tumor resection,<br />

alcohol ablation, radi<strong>of</strong>requency ablation, chemoembolization, radioembolization,<br />

chemotherapy or targeted therapy (Sorafenib).<br />

The approach for the management <strong>of</strong> HCC must be multi disciplinary where<br />

oncology, hepatology, liver transplantation, liver surgery, interventional radiology,<br />

and palliative care specialists are involved.<br />

Prevention<br />

Vaccination programs<br />

Vaccination is a very powerful measure to reduce the infection rate with hepatitis<br />

B and hence reduce the incidence <strong>of</strong> HCC. The nationwide hepatitis B vaccination<br />

program launched in Taiwan in 1984 led to a reduction <strong>of</strong> the hepatitis carrier rate<br />

in children from 10% to less than 1% and to a reduction in the incidence <strong>of</strong> HCC<br />

from 0.70 to 0.36 per 100,000 between 1986 and 1994. In Saudi <strong>Arab</strong>ia, a routine<br />

hepatitis B vaccination <strong>of</strong> children was added as part <strong>of</strong> the extended program<br />

<strong>of</strong> immunization in 1989. A dramatic reduction was noted in the prevalence <strong>of</strong><br />

hepatitis B from 6.7% in 1989 to 0.3% in 1997. No evidence is available yet on<br />

the effect <strong>of</strong> this reduction on the incidence <strong>of</strong> HCC.<br />

Treatment <strong>of</strong> viral hepatitis<br />

If cirrhosis is the most important risk factor for the development <strong>of</strong> HCC, could<br />

the incidence <strong>of</strong> HCC be reduced by preventing cirrhosis or treating cirrhosis due<br />

to viral hepatitis with antiviral therapy? Many studies in hepatitis B and hepatitis<br />

C show that treatment <strong>of</strong> active hepatitis, especially when successful, may lead<br />

to a reduction in the incidence <strong>of</strong> HCC.<br />

Screening<br />

Although many modeling data exist suggesting that screening can reduce HCC<br />

related disease specific mortality in a cost effective manner, there is only one<br />

randomized trial showing benefit while many other studies didn’t. In the study<br />

showing benefit from China, 18,816 patients where screened with 6 monthly<br />

AFP and ultrasound showing a reduced mortality rate by 37% in the screened<br />

arm even though the adherence to the surveillance was only around 60%. The<br />

screened population in this study was patients with current or previous exposure<br />

to hepatitis B.<br />

Patients who are best candidates for enrollment in a screening program include<br />

patients with cirrhosis for whatever reason. Patients who have hepatitis B without<br />

cirrhosis, especially if above 45 in age or have a family history <strong>of</strong> HCC may also<br />

be candidates.<br />

Most authorities (including the Saudi Gastroeterology <strong>Association</strong>) suggest<br />

screening with abdominal ultrasound and AFP every 6 months. Once a lesion is<br />

detected then a triphasic CT or contrast enhanced MRI is indicated for confirmation.<br />

In case <strong>of</strong> patients with cirrhosis who have a lesion larger than 2 cm detected by a<br />

contrast radiological study showing early arterial enhancement and rapid venous<br />

washout this is diagnostic for HCC and no further testing is recommended. If the<br />

lesion is less than 2 cm or the patient is not cirrhotic then a biopsy is indicated<br />

for confirmation.<br />

Outcome<br />

The natural history <strong>of</strong> HCC depends on the stage <strong>of</strong> the disease but is poor in the<br />

majority <strong>of</strong> cases. Tumor size at presentation is an important factor in the natural<br />

history but its use as a sole predicting factor is hindered by the fact that this tumor<br />

doubling time may in fact be very variable. In some patients the tumor growth is<br />

slow doubling in size in twenty months or more, while in others the tumor grows<br />

much faster and doubles in less that 1 month. In symptomatic patients in China<br />

and Africa, death usually occurs within four months, while some reports suggest<br />

a longer survival and a more indolent course in Western countries. The most<br />

important factor in determining the natural history <strong>of</strong> HCC patients is the stage<br />

<strong>of</strong> the underlying liver disease.<br />

Recommendation <strong>of</strong> Hepatocellular Carcinoma Prevention and Early Detection<br />

Population Category Recommendation<br />

Primary Prevention General population - Hep B vaccination<br />

- Prevention <strong>of</strong> hepatitis<br />

- Avoidance <strong>of</strong> alcohol<br />

Patients with hepatitis - Treatment <strong>of</strong> hepatitis<br />

Patients with cirrhosis Early detection Alpha feto protein and<br />

liver ultrasound every 6<br />

months<br />

References<br />

1. J Bruix, M Sherman. Management <strong>of</strong> hepatocellular carcinoma. Hepatology<br />

2005; 42(5):1208-1236.<br />

2. Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia ministry <strong>of</strong> health, National Cancer Registry (1997-<br />

1998). 2001.<br />

3. Llovet JM, Bruix J. Systematic review <strong>of</strong> randomized trials for unresectable<br />

hepatocellular carcinoma: Chemoembolization improves survival. Hepatology<br />

2003; 37(2):429-442.<br />

4. Chang M, Chen C, Lai M, Hsu H, Wu T, King M. Universal hepatitis B vaccination<br />

in Taiwan and the incidence <strong>of</strong> hepatocellular carcinoma in children. N Engl J<br />

Med 1997; 336:1855-1859.<br />

5. Liaw Y, Sung J, Chow C, Farrell G, Lee C, Yuen H et al. Lamivudine for<br />

patients with chronic hepatitis B and advanced liver disease. N Engl J Med 2004;<br />

35:1521-1531.<br />

6. Zhang B, Yang B, Tang Z. Randomized controlled trial <strong>of</strong> screening for<br />

hepatocellular carcinoma. J Cancer Res Clin Oncol 2004; 130:417-422.<br />

7. Abdo A et al. Saudi Gastroenterology <strong>Association</strong> guidelines for the diagnosis<br />

and management <strong>of</strong> hepatocellular carcinoma, summary <strong>of</strong> recommendations.<br />

Annls Saudi Med 2006, 26: 261-265<br />

8. Llovet J et al. Sorafoneb in advanced hepatocellular carcinoma. NEJM<br />

2008,359:378-90<br />

78 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


CANCER PREVENTION AND EARLY DETECTION IN CHILDREN<br />

MALIGNANCIES<br />

Reem Al Sudairy, MD<br />

King Abdulaziz <strong>Medical</strong> City for National Guard, Riyadh, KSA<br />

Corresponding Author: Reem Al Sudairy, MD<br />

Department <strong>of</strong> <strong>Oncology</strong> (Mail Code 1777)<br />

P.O. Box 22490, Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

E-mail: sudairyr@ngha.med.sa<br />

Abstract<br />

Objectives: Cancer in children is rare disease. It accounts for about 1% <strong>of</strong> all<br />

malignancies, and as a result, understanding <strong>of</strong> the factors causing childhood<br />

cancer is less well defined compared to that for adults. <strong>Medical</strong> research has<br />

contributed greatly to improved treatment outcome in childhood cancer, reaching<br />

cure rates up to 80%. However, little progress has been made in prevention <strong>of</strong><br />

childhood cancer. This manuscript summarizes the early detection and prevention<br />

guidelines for children.<br />

Methods: Review <strong>of</strong> the literature and international pediatric oncology<br />

recommendations about childhood cancer prevention and early detection are<br />

reviewed and summarized.<br />

Results: Genetic predisposition can cause up to 10% <strong>of</strong> all childhood malignancies.<br />

This important risk factor will be discussed in details with emphasis on<br />

recommendations for genetic testing and follow up <strong>of</strong> those children by the<br />

primary care physicians.<br />

Environmental / external factors in relation to childhood cancer will be also<br />

reviewed with updated information on current research pertinent to this important<br />

risk factor. Recommendations to pediatricians and primary care physicians<br />

concerning early detection and prevention <strong>of</strong> childhood malignancies in at risk<br />

population will be also discussed.<br />

While there is still a knowledge gap in the field, there are certain clinical situation<br />

where early detection and prevention will help in the control <strong>of</strong> childhood cancer.<br />

Introduction<br />

Cancer in children is rare and accounts for about 1% <strong>of</strong> all malignancies and as a<br />

result the precise causes <strong>of</strong> childhood cancers are still insufficiently known, and they<br />

are less well defined compared to that for adults. Childhood Cancers is typically<br />

<strong>of</strong> different variety from those observed in adults. The carcinogenic process in<br />

children is much shorter in time. In children cancers are mainly mesenchymal /<br />

neural in origin while in adults they are mainly epithelial and in internal organs<br />

and have a strong and proven link to environmental factors.<br />

Incidence<br />

Approximately 149 <strong>of</strong> every 1 million children under the age <strong>of</strong> 20 years are<br />

diagnosed with cancer each year in the United States. Acute leukemia accounts<br />

for the greatest proportion <strong>of</strong> new cancer cases (25-30%) followed by brain tumors<br />

(20%) and lymphomas (15%). (1)<br />

According to the Saudi Cancer Registry 2004 statistics, the total incident cases<br />

reported among children (0-14 years) were 713 which represent 7.6 % <strong>of</strong> the<br />

total number <strong>of</strong> cancers in Saudi <strong>Arab</strong>ia. Of all the cases reported, there were<br />

584 Saudis. (2)<br />

Although the incidence <strong>of</strong> pediatric cancer is low, its significance is <strong>of</strong> great<br />

importance. In KSA in 2007 approximately 40% <strong>of</strong> the Saudi population was under<br />

15 years which put a large number <strong>of</strong> the population at risk <strong>of</strong> childhood cancer.<br />

Childhood Cancer Symptoms<br />

The symptoms <strong>of</strong> childhood cancer depend on the site and the extent <strong>of</strong> the tumor.<br />

Leukemia can cause anemia, bleeding, fever, bone pain and lymph nodes, spleen,<br />

liver enlargement. Most <strong>of</strong> other tumors produce symptoms related to their position<br />

either in the form <strong>of</strong> a lump or because it impairs the function <strong>of</strong> one or more organs.<br />

Most <strong>of</strong> children with cancer are treated at pediatric cancer centers per national<br />

clinical protocols. Surgery, chemotherapy, radiotherapy are the mainstay <strong>of</strong><br />

treatment for most childhood cancers. Bone marrow and peripheral stem cell<br />

transplantation is another important modality <strong>of</strong> therapy in some cases.<br />

Outcome<br />

Although cancer still represents the second most common cause <strong>of</strong> death in children<br />

(following accidents), the survival rate from children cancer have improved<br />

substantially in the last 30 years and had risen to over 75% due to improvement in<br />

treatment modalities. There is a relative greater improvement in the young age group<br />

(under 15 years) in comparison to older children (15-19 years <strong>of</strong> age). Although<br />

disease biology may play a role in this difference in outcome, failure to treat<br />

older children on national protocols plays a major role in their inferior outcome.<br />

Risk Factors<br />

The precise causes <strong>of</strong> many childhood cancers are still not well understood but<br />

they are assumed to be multi-factorial. The two most important risk factors are:<br />

1. Genetic predisposition.<br />

2. Environmental factors (Inutero and during early childhood).<br />

Genetic pre-disposition to Cancer<br />

A large number <strong>of</strong> predisposing syndromes exist and account for up to 10% <strong>of</strong><br />

all childhood malignancies. Most syndromes are associated with a germ line<br />

mutation in a single gene (e.g. RB1) however; in some syndromes (e.g. Wilm’s<br />

Tumor) several genetic loci have been involved. Polymorphism <strong>of</strong> certain genes<br />

loci have been shown to play a role in cancer development. (3)<br />

Table 1 shows the genetic conditions predisposing to cancer in children. It is<br />

the responsibility <strong>of</strong> the pediatricians/family physician’s to be able to recognize<br />

clinically cancer-predisposing syndromes, and should strongly suspect the presence<br />

<strong>of</strong> cancer-predisposing condition from the family history (earlier age <strong>of</strong> cancer<br />

onset, bilateral or multifocal tumors, and multiple primary malignancies <strong>of</strong><br />

different types in the same individuals). The presence <strong>of</strong> certain physical signs in<br />

parent should also alert physicians, like café-au-lait spots and axillary freckling<br />

(Neur<strong>of</strong>ibromatosis type1). All at risk children should be referred to a trained<br />

genetic counselor and should be followed up regularly by their pediatrician/<br />

family physician.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 79


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

Table 1: The genetic conditions predisposing to cancer in children.<br />

No. Disease Inheritance Most common cancers<br />

1. Ataxia telangiectasia Recessive Leukemia, Lymphoma<br />

2. Beckwith-Wiedemann syndrome Complex Wilms’ Tumor, Hepatoblastoma, Adrenal carcinoma.<br />

3. Bloom’s Syndrome Recessive Leukemia, Lymphoma, Epithelial Cancers, Hepatocellular Carcinoma,<br />

Sarcomas, Brain Tumor.<br />

4. Familial Adenomatous Polyposis Dominant Colorectal Carcinoma, Hepatoblastoma, Thyroid Cancer, Medulloblastoma.<br />

5. Fanconi Anemia Recessive Leukemia, Squamous Cell carcinoma and Gastrointestinal and<br />

Genitourinary Tract Tumors.<br />

6. Juvenile Polyposis Dominant Gastrointestinal tumors<br />

7. Li-Fraumeni Syndrome Dominant S<strong>of</strong>t tissue sarcomas,Osteosarcoma, Breast cancer, Brain tumors,<br />

Adrenocortical carcinoma, Leukemia.<br />

8. Multiple endocrine neoplasia, type 1 Dominant <strong>Pan</strong>creatic islet cell tumor, Pituitary and Parathyroid adenoma.<br />

9. Multiple endocrine neoplasia, type 2 Dominant Medullary thyroid carcinoma, Pheochromocytoma, Parathyroid hyperplasia.<br />

10. Neur<strong>of</strong>ibromatosis, type 1 Dominant Neur<strong>of</strong>ibromas, Optic pathway gliomas, Leukemia, Malignant peripheral<br />

nerve sheath tumors.<br />

11. Neur<strong>of</strong>ibromatosis, type 2 Dominant Vestibular Schwannoma<br />

12. Nijmegen breakage syndrome Recessive Lymphoma, Medulloblastoma, Glioma.<br />

13. Nevoid basal cell carcinoma syndrome Dominant Medulloblastoma, basal cell carcinomas.<br />

14. Peutz-Jeghers syndrome Dominant Intestinal tumors, Gastric and <strong>Pan</strong>creatic canceras, Gonadal tumors.<br />

15. Retinoblastoma Dominant Retinoblastoma, Osteosacoma.<br />

16. Rhabdoid predisposition syndrome Dominant Rhabdoid tumor, Medulloblastoma, choroid plexus tumor<br />

17. Rothmund Thomson Syndrome Recessive Skin and Bone tumors.<br />

18. Simpson Golabi Behmel Syndrome X-linked Recessive Wilms’ Tumor, Hepatoblastoma.<br />

19. Sotos syndrome Dominant Sacrococcygeal teratoma, Neuroblastoma, Leukemia, Lymphoma, Wilms’<br />

Tumor<br />

20. Tuberous Sclerosis Dominant Central Nervous System tumors, Hamartomas, Renal angiomyolipoma,<br />

Renal cell carcinoma.<br />

21. Von Hippel-Lindau syndrome Dominant Retinal and Central Nervous System Hemangioblastoma,<br />

Pheochromocytoma, Renal cell carcinoma.<br />

22. Werner Syndrome Recessive Osteosarcoma, Menigioma, Melanoma, Thyroid carcinoma.<br />

23. Wilm’s Tumor Syndromes Dominant Wilms’ Tumor.<br />

24. Xeroderma pigmentosum Recessive Skin cancer, Leukemia<br />

25. X-linked lymphoproliferative disease X-linked Recessive Lymphoma<br />

Genetic testing is not a standard <strong>of</strong> care for all <strong>of</strong> the syndromes associated with<br />

pediatric malignancies, but in some <strong>of</strong> these syndromes where there is an increase<br />

risk <strong>of</strong> development <strong>of</strong> cancer during childhood; there is increasing evidence in<br />

the literature that using genetic testing and cancer surveillance has enhanced the<br />

long term outcome for affected patients. (3, 4, 5)<br />

American Society <strong>of</strong> Clinical <strong>Oncology</strong> (ASCO) issued a statement update on<br />

genetic testing for cancer susceptibility in 2003. The following are some <strong>of</strong> the<br />

recommendations that were made: (6)<br />

1-Indications for genetic testing:<br />

• The individuals have personal or family history features suggestive <strong>of</strong> a<br />

genetic cancer susceptibility condition.<br />

• The test can be adequately interpreted.<br />

• The results will aid in diagnosis or influence the medical or surgical<br />

management <strong>of</strong> the patient <strong>of</strong> family members at hereditary risk <strong>of</strong> cancers.<br />

2-Special issues in testing children for cancer susceptibility<br />

• ASCO recommends that the decision to <strong>of</strong>fer testing to potentially affected<br />

children should take into account the availability <strong>of</strong> evidence-based riskreduction<br />

strategies and the probability <strong>of</strong> developing a malignancy during<br />

childhood.<br />

• Where risk-reduction strategies are available or cancer predominantly<br />

develops in childhood, ASCO believes that the scope <strong>of</strong> parental authority<br />

encompasses the right to decide for or against testing.<br />

• In the absence <strong>of</strong> increased risk <strong>of</strong> childhood malignancy, ASCO recommends<br />

delaying genetic testing until an individual is <strong>of</strong> sufficient age to make an<br />

informed decision regarding such tests.<br />

• The clinical cancer genetics pr<strong>of</strong>essional should be advocate for the best<br />

interests <strong>of</strong> the child.<br />

3-Counseling pre- and post-test:<br />

ASCO recommends that genetic testing only be done in setting <strong>of</strong> pre and posttest<br />

counseling, which should include discussion <strong>of</strong> possible risks and benefits <strong>of</strong><br />

cancer early detection and prevention modalities.<br />

80 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


Tumor Surveillance for Children with Cancer Predisposing Syndromes<br />

Unfortunately in children (in contrast to adults), only few guidelines for surveillance<br />

have been established and their benefit has been proven. The following are<br />

some examples <strong>of</strong> inherited cancer predisposing syndromes that have established<br />

pragmatic surveillance guidelines that are based on available evidence from large<br />

studies.<br />

Retinoblastoma (RB)<br />

It is a malignant tumor <strong>of</strong> the embryonic cells <strong>of</strong> the retina. It accounts for up to 4%<br />

<strong>of</strong> all childhood cancers. It occurs in heritable form (40%) where it will be usually<br />

bilateral or multifocal in younger patients (median age 11 months), and it occurs in<br />

nonheritable form (60%) where it will be unilateral in older patients (median age <strong>of</strong><br />

22 months). Approximately 90% <strong>of</strong> RB cases are diagnosed by the age <strong>of</strong> 5 years.<br />

• It is recommended that children at risk <strong>of</strong> RB (including those with germ line<br />

RB1 gene mutation, as well as their <strong>of</strong>fspring or siblings) should undergo<br />

ophthalmologic examination under general anaesthesia starting at birth.<br />

Recommendation for screening intervals and length <strong>of</strong> follow-up vary, but<br />

most experts recommend frequency <strong>of</strong> every 2-3 months until the age <strong>of</strong> 2<br />

years by which time 90% <strong>of</strong> heritable RB occur, after that frequency will be<br />

spaced to every 4-6 months until fourth to fifth year <strong>of</strong> life. (3,4,7)<br />

• To screen for pineal tumors, children should undergo MRI examinations <strong>of</strong><br />

the brain every 6 months until the age <strong>of</strong> 5 years. (4,8)<br />

• No standard screening protocols for secondary tumors.<br />

Beckwith-Wiedmann Syndrome (BWS) and Idiopathic Hemihypertrophy<br />

(IHH)<br />

BWS is a congenital disorder <strong>of</strong> growth regulation characterized by macroglossia,<br />

macrosomia, ear anomalies, abdominal wall defects and neonatal hypoglycemia.<br />

In patients with BWS/IHH tumor surveillance is recommended. It should be<br />

targeted at Wilms’ tumor (the commonest childhood tumor <strong>of</strong> the kidney) and<br />

Hepatoblastoma. (9, 4, 10)<br />

The following should be performed on regular intervals:<br />

• Renal ultrasound, every 3 months until the age <strong>of</strong> 8 years.<br />

• Liver ultrasound, every 3 months until the age <strong>of</strong> 4 years.<br />

• Measurements <strong>of</strong> serum alpha feto protein (AFP) every 3 months until the<br />

age <strong>of</strong> 4 years.<br />

Wilms’ Tumor Associated Syndromes<br />

There are several genetic syndromes other than BWS/IHH which might predispose<br />

to Wilms’ tumor like WAGR syndrome, Denys-Drash syndrome, Familial Wilms’<br />

tumor, Perlman syndrome, Fanconi anemia, Frasier syndrome and Simpson-Golabi-<br />

Behmel syndrome. Screening by ultrasound abdomen should be performed to<br />

those patients every 3 – 6 months as they have > 5% risk <strong>of</strong> Wilms' tumor (10,11)<br />

until the age <strong>of</strong> 5 years as it will detect 90-95% <strong>of</strong> tumors in patients with WT1<br />

gene mutation (10,4).<br />

Multiple Endocrine Neoplasia (MEN disorders)<br />

MEN disorders are cancer family syndromes that affect different endocrine organs.<br />

• MEN1 mutation carriers should be screened by annual serum calcium,<br />

parathyroid hormones, fasting glucose, insulin, prolactin and insulin growth<br />

factor 1. They should also have MRI examinations <strong>of</strong> the brain to assess the<br />

anterior pituitary gland every 3 years. (5)<br />

• MEN2 mutation carriers, all children should receive a prophylactic<br />

thyroidectomy early in life. (3,5)<br />

Familial Adenomatous Polyposis (FAP)<br />

FAP mutation carriers should be screened by annual colonoscopy beginning<br />

between the age <strong>of</strong> 10 and 14 years. If Polyps are found, colectomy should be<br />

performed using modern surgical techniques that preserve fecal continence. (3,<br />

12, 13)<br />

Von Hippel-Landau Disease (VHL)<br />

VHL is characterized by the development <strong>of</strong> proliferation <strong>of</strong> blood vessels<br />

in the retina, cerebellum or the spinal cord. It is characterized by cerebellar<br />

hemangioblastoma, retinal angioma, renal cell carcinoma and pheochromocytoma<br />

and other visceral tumors. Diagnosis usually occurs during early adulthood.<br />

Based on National Institute <strong>of</strong> Health-USA recommendations, patients and<br />

individuals at risk <strong>of</strong> VHL should receive an annual ophthalmologic examination<br />

from 1 year <strong>of</strong> age, annual urine catecholamine measurements from 2 years <strong>of</strong><br />

age, enhanced MRI examination <strong>of</strong> the brain and spine every 2 years starting at 11<br />

years <strong>of</strong> age and then every 3-5 years after the age <strong>of</strong> 60 years, annual abdominal<br />

ultrasound beginning at 11 years <strong>of</strong> age, and abdominal CT scan every 1-2 years<br />

after the age <strong>of</strong> 20 years. Additionally, regular complete physical examination<br />

is suggested for detection <strong>of</strong> early abnormal cerebellar and spinal cord signs.<br />

(3, 14, 15)<br />

For the other cancer predisposing syndromes, there are no clear, widely used,<br />

standard surveillance protocols in the literatures.<br />

Environmental/External Factors<br />

Cancers are assumed to be multi-factorial diseases that occur when a complex<br />

and prolonged process involving genetic and environmental factors interact in a<br />

multistage sequence. In the last few decades, environmental linkages have been<br />

actively investigated and described, but the evidence for causal association is still<br />

in the primary stages especially in the field pediatric oncology. Etiological studies<br />

have <strong>of</strong>ten been concerned with exposures occurring during the mother’s pregnancy<br />

although pre-conception and post natal factors have also been investigated.<br />

Numerous environmental exposures have been linked with childhood cancer.<br />

Interpretation is limited by study designs (retrospective, case control studies),<br />

variation in the timing <strong>of</strong> exposure ranging from before conception to during the<br />

child’s lifetime, the wide range <strong>of</strong> cancers studied and most importantly the small<br />

numbers <strong>of</strong> patients affected with such a rare disease.<br />

In 2004 International Childhood Cancer Cohort Consortium (I4C) was proposed<br />

and since then it has progressed steadily. The purpose <strong>of</strong> the Consortium is<br />

the prevention <strong>of</strong> childhood cancer using evidence from prospective children’s<br />

cohort studies around the world. They are trying to advance understanding <strong>of</strong><br />

the etiology and carcinogenic mechanisms in relation to childhood cancer. The<br />

study has 11 participating cohorts form 9 countries around the world with about<br />

700,000 subjects. (16, 17)<br />

Antenatal priority domains <strong>of</strong> interest for I4C outcomes are parental occupation/<br />

type <strong>of</strong> work, smoking/drug use (mother/father, passive (maternal)), diet (fish,<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 81


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

seafood and yogurt), radiation exposure, pesticide/chemical exposure, maternal<br />

infection, sun exposure/Vitamin D, supplements during pregnancy and folate intake.<br />

For children, postnatal priority domains include: Anthropometry, infectious up to<br />

1 year, radiation exposure, feeding habits, sun exposure, passive smoking, atopy<br />

/asthma. The reader is referred to an excellent review on environmental linkages<br />

for childhood cancer. (18)<br />

From that review, environmental factors that had a conclusive evidence (cause<br />

and effect are undoubtedly linked with evidence <strong>of</strong> a dose-response trend) were :<br />

high-dose ionizing radiation linked to thyroid cancer, prenatal diethylstilboestrol<br />

linked to vaginal adenocarcinoma and tobacco smoke linked to lung cancer<br />

during adulthood.<br />

Other environmental factors with compelling evidence (substantial data linking<br />

cause and effect but with no consistent evidence <strong>of</strong> a dose-response trend and/or<br />

evidence confirming timing and dosing <strong>of</strong> exposure) were: diagnostic radiographs,<br />

industrial air pollution, solar UV radiation and viral agents.<br />

There was inconclusive evidence (extensive studies have produced inconsistent<br />

results) that other environmental factors were linked to childhood cancer like:<br />

residential proximity to nuclear facilities, extremely low-frequency electromagnetic<br />

field (ELF-EMF), traffic-related air pollution, hot dogs and parental occupational<br />

exposures. Until conclusive evidence is available on environmental factors it is<br />

recommended that: Parents should avoid exposure to substances that are known<br />

or suspected carcinogens, and this advice holds during pregnancy and at all<br />

stages <strong>of</strong> life. Behaviors that might enhance protection against cancer in <strong>of</strong>fspring<br />

include taking multivitamin supplements during pregnancy, breast feeding as long<br />

as possible. Children should avoid exposure to various carcinogens. Instilling<br />

healthy lifestyle choices such as optimizing the child’s dietary intake <strong>of</strong> fibers,<br />

fruits and vegetables, exercise and dietary moderation during childhood lays an<br />

important foundation for long-term cancer prevention. Focusing on children and<br />

young adolescents in primary prevention is very important as it is easier to teach<br />

healthy behaviors at a young age rather than modify behaviors at later age. (19, 20)<br />

Physicians should avoid performing unnecessary diagnostic radiographs.<br />

Long Term Follow-up Guidelines for Survivors <strong>of</strong> Childhood, Adolescent<br />

and Young Adult Cancers<br />

The improvement in survival rates in children with cancer resulted in a growing<br />

population <strong>of</strong> childhood cancer survivors. The use <strong>of</strong> cancer therapy at an early<br />

age can produce long term complications, such as impairment in growth and<br />

development, neurocognitive deficits, cardiac, pulmonary, gastrointestinal, renal,<br />

endocrine and gonodel dysfunction as well as second malignant neoplasms. (21)<br />

Several studies following up large cohorts <strong>of</strong> survivors <strong>of</strong> childhood cancers<br />

reported 3-10 fold increased risk for second malignancies in comparison with<br />

general population, which are a leading cause <strong>of</strong> non-relapse- related late mortality.<br />

(22)<br />

Second malignancies in childhood survivors vary depending on the type <strong>of</strong><br />

therapy received and the presence <strong>of</strong> genetic predisposition. They are classified<br />

into two groups: (22)<br />

• Therapy-related myelodysplasia/acute myeloid leukemia, which is<br />

characterized by short latency (approximately 3-5 years from primary<br />

cancer diagnosis) and exposure to alkylating agents and/or topoisomerase<br />

II inhibitors.<br />

• Therapy-related solid tumors, which has a strong and well-defined association<br />

with radiation and characterized by latencies that exceed 10 years.<br />

Guidelines For Screening Childhood Cancer Survivors: The Children’s<br />

<strong>Oncology</strong> Group (COG) developed risk-based exposure related clinical practice<br />

guidelines[Long-Term Follow-Up Guidelines for survivors <strong>of</strong> childhood,<br />

adolescent, and young adult cancers (COG-LTFU) ] for screening and management<br />

<strong>of</strong> late effects resulting from therapeutic exposures used during treatment for<br />

pediatric malignancies.<br />

Those guidelines were developed collaboratively by the COG Nursing Discipline<br />

and Late Effects Committee. They represent a statement <strong>of</strong> consensus from a panel<br />

<strong>of</strong> experts in late effects <strong>of</strong> pediatric cancer treatment. The recommendations<br />

are based on thorough review <strong>of</strong> the literatures as well as the collective clinical<br />

experience <strong>of</strong> the taskforce members, panel <strong>of</strong> experts and multidisciplinary<br />

review panel (including nurses, physicians, behavioral specialists and patient/<br />

parent advocates).<br />

The Guidelines and the Health Links can be downloaded from www.survivorship.<br />

guidelines.org<br />

Table 2: Selected exposure-based screening recommendations (Children’s<br />

<strong>Oncology</strong> Group Long-Term Follow-Up (COG-LTFU) Guidelines).<br />

Category Therapeutic Potential Late Recommended<br />

Exposure Effect Screening<br />

Second Etoposide Acute Myeloid CBC, platelet,<br />

Malignancies Teniposide Leukemia differential yearly for<br />

10 years following<br />

exposure<br />

Alkylating Acute Myeloid CBC, platelet,<br />

Chemotherapy Leukemia/ differential yearly for<br />

Anthracyclines Myelodysplasia 15 years following<br />

exposure<br />

Radiation (any Second Yearly history and<br />

field) Malignant physical exam with<br />

neoplasm inspection and<br />

(SMN) in palpalation <strong>of</strong> tissues in<br />

radiation field<br />

(skin, bone,<br />

s<strong>of</strong>t tissue)<br />

radiation field.<br />

Radiation<br />

impacting the<br />

thyroid<br />

Thyroid Cancer Yearly thyroid exam<br />

Radiation Breast Cancer Monthly breast exam<br />

impacting the<br />

Clinician breast exam<br />

breast<br />

yearly until age 25, then<br />

every 6 months<br />

Mammogram yearly<br />

beginning 8 years after<br />

radiation or at age 25,<br />

whichever comes last.<br />

Radiation Colorectal Colonoscopy every 10<br />

impacting the Cancer years beginning 10 years<br />

colon<br />

following radiation or<br />

at age 35, whichever<br />

comes last<br />

82 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


Pediatric Cancer Prevention and Early Detection Guidelines<br />

Intervention Population Procedure Frequency Starting Age Stopping Age<br />

Primary Parents Avoid exposure to substances that are known or N/A N/A N/A<br />

Prevention<br />

suspected carcinogens<br />

Multivitamins supplement during pregnancy N/A N/A N/A<br />

Breast feeding as long as possible N/A N/A N/A<br />

Children Avoid exposure to various carcinogens N/A N/A N/A<br />

Instilling healthy lifestyle choices N/A N/A N/A<br />

Physicians Avoid performing unnecessary diagnostic<br />

radiographs.<br />

N/A N/A N/A<br />

Screening General Public<br />

Special Population<br />

N/A N/A N/A N/A<br />

Screening Retinoblastoma (RB) 1. Opthalmologic examination under general<br />

anaesthesia<br />

every 2-3 months<br />

till age <strong>of</strong> 2 years<br />

then every 6<br />

months<br />

Till age <strong>of</strong> 4 years<br />

at birth 4-5 years<br />

2. Screen for pineal tumors, do MRI brain Every 6 months At birth 5 years<br />

Beckwith-Wiedmann Syndrome and<br />

Idiopathic Hemihypertrophy (IHH)<br />

1. Renal ultrasound every 3 months Diagnosis 8 years<br />

2. Liver ultrasound every 3 months Diagnosis 4 years<br />

3. Measurement <strong>of</strong> serum AFP every 3 months Diagnosis 4 years<br />

Wilms' Tumor Associated Syndromes 1. Abdominal Ultrasound every 3-6 months Diagnosis 5 years<br />

Multiple Endocrine Neoplasia (MEN<br />

Disorders)<br />

MEN 1 Mutation Carriers<br />

1. Serum Calcium, parathyroid hormones,<br />

fasting glucose, insulin, prolactin and insulin<br />

growth factor 1.<br />

every year Diagnosis indefinite<br />

2. MRI examinations <strong>of</strong> the brain to assess the<br />

anterior pituitary gland.<br />

MEN 2 Mutation Carriers<br />

every 3 years Diagnosis indefinite<br />

1. Prophylactic thyroidectomy early in life N/A N/A N/A<br />

Familial Adenonatous Polyposis<br />

(FAP)<br />

1. Colonoscopy every year 10-14 years indefinite<br />

Von Hippel-Landau Disease (VHL) 1. Ophthalmologic examination every year 1 year <strong>of</strong> age indefinite<br />

2. Urine Catecholamine measurements every year 2 years <strong>of</strong> age indefinite<br />

3. MRI examination <strong>of</strong> the brain and spine every 2 years 11 years <strong>of</strong><br />

age<br />

60 years<br />

every 3-5 years 60 years indefinite<br />

Survivors <strong>of</strong> childhood Cancer Refer to Table 2 for details <strong>of</strong> screening recommendations<br />

References<br />

1. Gurney JG, Bondy ML. Epidemiology <strong>of</strong> pediatric oncology. In: Pizzo P,<br />

Poplack D, editors. Principles and practice <strong>of</strong> pediatric oncology. 6th edition.<br />

Philadelphia: Lippincott, Williams and Wilkins; 2006. P. 1-13.<br />

2. Saudi Cancer Registry, Cancer incidence Report, 2004. Page 24.<br />

3. Pakakasama S, Tomlinson GE. Genetic predisposition and screening in<br />

pediatric cancer. Pediatric Clinic <strong>of</strong> North America-volume 49, issue 6(Dec.<br />

2002):1393-1413.<br />

4. Rao A, Rothman J, Nichols KE. Genetic testing and tumor surveillance for children<br />

with cancer predisposition syndromes. Current Opinion in pediatrics 2008, 20:1-7.<br />

5. Strahm B, Malkin D, Hereditary cancer predisposition in children: Genetic<br />

basis and clinical implications. Int J Cancer 2006; 119:2001-2006.<br />

6. American Society <strong>of</strong> Clinical <strong>Oncology</strong> policy statement update: Genetic<br />

testing for cancer susceptibility. J Clin Oncol 2003; 21:2397-2406.<br />

7. Moll AC, Imh<strong>of</strong> SM,Schouten-Van Meeteren YN, et al.At what age could<br />

screening for familial retinoblastoma be stopped? A register based study 1945-<br />

98. Br J Ophthalmol 2000; 84:1170-2.<br />

8. Kivela T. Trilateral retinoblastoma: a meta-analysis <strong>of</strong> hereditary<br />

retinoblastoma associated with primary ectopic intracranial retinoblastoma. J<br />

Clin Oncol 1999; 17:1829-1837.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 83


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

9. Tan TY, Amor DJ. Tumor surveillance in Beckwith-Wiedemann syndrome and<br />

Hemihyperplasia: A critical review <strong>of</strong> the evidence and suggested guidelines for<br />

local practice. J Pediatr Child Health 2006; 42:486-490.<br />

10. Scott RH, walker L, Olsen OE,et al. Surveillance for Wilms' tumor in at-risk<br />

children: pragmatic recommendations for best practice. Arch Dis Child 2006;<br />

91:995-999.<br />

11. Scott RH, Stiller CA, Walker L, et al. Syndroms and constitutional<br />

chromosomal abnormalities associated with Wilms' tumor. J Med Genet 2006;<br />

43:705-715.<br />

12. Hyer W, Fell JM. Screening for familial adenomatouse polyposis. Arch Dis<br />

Child 2001;84:377-80.<br />

13. Plon SE, Malkin D. Childhood cancer and heredity. In: Pizzo P, Poplack D,<br />

editors. Principles and practice <strong>of</strong> pediatric oncology. 6th edition. Philadelphia:<br />

Lippincott, Williams and Wilkins; 2006:24.<br />

14. Plon SE, Malkin D. Childhood cancer and heredity. In: Pizzo P, Poplack D,<br />

editors. Principles and practice <strong>of</strong> pediatric oncology. 6th edition. Philadelphia:<br />

Lippincott, Williams and Wilkins; 2006:27-28.<br />

15. Singh AD, Shields CL, Shields JA. Von Hippel-Lindau disease. Surv<br />

Ophthalmol 2001; 46:117-42.<br />

16. Brown RC, Dwyer T, Kasten C, ET al.Cohort pr<strong>of</strong>ile: The International<br />

Childhood Cancer Cohort Consortium (I4C). Int J Epidemiol 2007; 36:724-730<br />

17. Second International Childhood Cancer Cohort Consortium<br />

Workshop. August 29-30, 2007.www.nationalchildrensstudy.gov/research/<br />

internationalinvolvement/pages/I4C-workshop-083007-101107.<br />

18. Buka I, Koranteng S, Vargas ARD. Trend in childhood cancer incidence:<br />

Review <strong>of</strong> Environmental Linkages. Pediatr Clin N Am 2007; 54:177-203.<br />

19. Pollock BH, Knudson Jr. AG. Preventing cancer in adulthood: Advise<br />

for pediatrician. In: Pizzo P, Poplack D, editors. Principles and practice <strong>of</strong><br />

pediatric oncology. 6th edition. Philadelphia: Lippincott, Williams and Wilkins;<br />

2006:1617-1628.<br />

20. Selvan MS, Kurpad AV. Primary prevention: why focus on children & young<br />

adolescents? Indian J Med Res 120, Dec. 2004, pp 511-518.<br />

21. Bhatia S. Cancer survivorship---Pediatric issues. American Society <strong>of</strong><br />

Hematology (ASH) educational book, 2005. P.507-515.<br />

22. Bhatia S. Secondary Malignancies: What, When, Why, in Whom? NCCN<br />

clinical practice oncology forum 2008. www.medcsape.com/viewarticle/581683<br />

ROLE OF PRIMARY CARE PHYSICIAN FIGHT AGAINST CANCER<br />

Farrah, M.<br />

Corresponding Author: Consultant Family Medicine & Primary Health Care<br />

Department <strong>of</strong> Family Medicine & Primary Health Care<br />

National Guard Health Affairs, King Abdulaziz <strong>Medical</strong> City<br />

P.O. Box 22490, Riyadh 11426, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

Abstract<br />

Background: Primary Care Physician (PCP) represents the first line in the fight<br />

against cancer for many reasons. Enhancing the roles <strong>of</strong> the PCPs will help in<br />

saving more lives from cancer related causes.<br />

Methods: The role <strong>of</strong> the PCP in the cancer prevention and early detection <strong>of</strong><br />

cancer were identified and summarized. The advantages <strong>of</strong> primary care physicians<br />

were enlisted.<br />

Results: PCP can play an important role in increasing the patient awareness<br />

and knowledge about cancer, help in decreasing the risks for preventable<br />

cancers and perform early detection for common cancers which will save lives.<br />

Recommendations for specific cancers are summarized.<br />

Conclusion: Enhancing the role <strong>of</strong> the primary care physicians in the fight<br />

against cancer is important due to certain advantages possess by them that can<br />

be better utilized. Physicians should incorporate cancer control activities into<br />

their routine practice.<br />

Introduction<br />

The structure <strong>of</strong> the current healthcare system is designed in such a way that it<br />

is largely dependent on the care seeking behavior <strong>of</strong> the patient, who initiates<br />

his or her care by the natural sensors in the body. A person would ask for help<br />

when a disease becomes symptomatic. Despite advances in technology and our<br />

knowledge that can detect some diseases even before symptoms appear, the care<br />

delivery system still lags behind in its metamorphosis. Even though radical changes<br />

are needed in the existing healthcare organizations to fully adapt to advances in<br />

knowledge, there is some overlap in the alignment <strong>of</strong> disease pathogenesis with<br />

the current levels <strong>of</strong> care.<br />

Primary care is where most diseases including cancer are first encountered. Care<br />

is further channeled to secondary and tertiary level for those who need it. The<br />

physicians working at the primary care have several advantages in applying some<br />

<strong>of</strong> the advances in technology and knowledge that relates to prevention and early<br />

detection <strong>of</strong> cancer. These advantages for the primary care physician are by default<br />

<strong>of</strong> existing design <strong>of</strong> care delivery system and with these come responsibilities<br />

that lie squarely on the primary care physicians.<br />

Advantages For Primary Care Physician<br />

First Contact<br />

The primary care physicians work at the frontline <strong>of</strong> any healthcare organization.<br />

These are either family medicine specialists or the non-specialist general<br />

practitioners. The primary care physicians work in solo or group practice and<br />

are mostly linked to tertiary care organizations. The primary care doctor is the<br />

first medical expert that the patient comes in contact with which allows for early<br />

detection <strong>of</strong> cancer.<br />

84 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


Opportunity<br />

Patients also seek counsel <strong>of</strong> their primary care physicians for problems other<br />

than cancer, this provides with the opportunity to screen for cancer early when<br />

it is not yet a problem for the patient to seek help for. Having a database <strong>of</strong> all<br />

normal patients who only seek help for smaller problems e.g. for an occasional<br />

upper respiratory tract infection, allows for the primary care physician to reach<br />

those with no symptoms <strong>of</strong> cancer by call-recall one by one.<br />

Wider Scope <strong>of</strong> Cancer Coverage<br />

The broader scope <strong>of</strong> work <strong>of</strong> the family physician allows them to cover screening<br />

for all screenable cancers, instead <strong>of</strong> being limited to cancer <strong>of</strong> one specialty or<br />

another.<br />

Multiplicity <strong>of</strong> Visits<br />

Patients have the most frequent interaction with their family physicians each<br />

year, than with any other specialty. Most cancer prevention screening procedures<br />

require no less than an annual follow-up.<br />

Family Network<br />

Being the family doctor for the whole family <strong>of</strong> the patient needing cancer<br />

prevention advice or screening, the primary care physician has several opportunities<br />

to alert and convince the family members to engage in cancer prevention screening<br />

for their other family members.<br />

Whole Family Coverage<br />

The visitors to family doctor are not limited by age or gender, therefore preventive<br />

coverage for cancers in all ages and genders can be initiated in the primary care<br />

setup. A person can be a patient <strong>of</strong> a family doctor for his or her lifetime. Family<br />

doctors <strong>of</strong>ten provide care to the next generation <strong>of</strong> their patients as well.<br />

Long Term Trust<br />

Having long-term relationship <strong>of</strong> trust with the patients and their families, the<br />

primary care physician is ideally placed to impart health education quickly, easily,<br />

with little resistance and with greater impact on the patient.<br />

Multi disciplinary Team Support<br />

The primary care physician's working team <strong>of</strong> nurses, health educators and others<br />

are as versatile as the scope <strong>of</strong> family practice to assist in managing the prevention<br />

<strong>of</strong> all cancers. Initiating the cancer screening for the patient is a simple process,<br />

based on age and gender and nursing staff at the primary care level have played<br />

a crucial role in several healthcare setups in this regard. Health educators do the<br />

counseling for cancer prevention where physicians are sometimes limited by<br />

their busy schedules.<br />

Coordination <strong>of</strong> Care<br />

The primary care physicians being in the role <strong>of</strong> classic gate-keeper, can coordinate<br />

the care <strong>of</strong> all types <strong>of</strong> cancers with the specialties that a patient may need referral<br />

to, thereby facilitating prompt treatment.<br />

Community-link<br />

The family doctors also have the most direct link with the community to which<br />

the patient belongs, which by default allows them to be in a leadership position to<br />

mobilize communities towards cancer prevention. Primary prevention for many<br />

cancers start with health promotion activities at the community level and no one<br />

is better suited than the family medicine physician for the job.<br />

Home-visits<br />

The family physicians and their team members are <strong>of</strong>ten the direct providers <strong>of</strong><br />

home health care, placing them at a key position in completing the loop <strong>of</strong> home<br />

based cancer care <strong>of</strong> tertiary prevention, i.e. rehabilitation and palliative care.<br />

Role Expectations In Fight <strong>Against</strong> Cancer<br />

Being in the position <strong>of</strong> several advantages within the existing healthcare structure,<br />

the primary care physician is expected to take the lead role in several aspects <strong>of</strong><br />

the fight against cancer, particularly in the arenas <strong>of</strong> primary prevention (health<br />

promotion and specific protection) and secondary prevention (screening). Family<br />

physicians are ideally positioned to implement clinical prevention guidelines for<br />

cancer. Their experience and feedback would be vital for the guidelines to have<br />

a successful pragmatic outcome. The roles <strong>of</strong> the primary care physicians are<br />

given below as general and specific as these relate to cancer. In addition the future<br />

roles and expectations are also described based on technological advances and<br />

anticipated changes in healthcare systems.<br />

General Role<br />

Guideline & Program Development<br />

Any guidelines developed or planning carried out at national, regional or local<br />

level would require the input <strong>of</strong> the representatives <strong>of</strong> family physician to develop<br />

and improve the implementation section <strong>of</strong> the guidelines or programs designed<br />

for cancer prevention.<br />

Primary Prevention<br />

Advocacy<br />

This role is not exclusive to the primary care physicians but they should not fall<br />

behind in advocating and raising awareness through news papers, magazines,<br />

television, radio and other mediums / forums <strong>of</strong> communicating with the public and<br />

the decision making bodies, such as government, non-governmental organizations,<br />

businesses and others who have the resources to bring change in healthcare delivery<br />

system and the attitudes about cancer<br />

Community Mobilization<br />

This requires that the primary care physician extends his/her influence as a<br />

healthcare pr<strong>of</strong>essional beyond the boundaries <strong>of</strong> the clinic and into the local<br />

community from which s/he draws practice. This measure is two-fold; first is<br />

recruiting <strong>of</strong> patients to form volunteer groups who believe in promoting healthier<br />

lifestyle in the community and also harnessing support <strong>of</strong> the local community<br />

leaders, followed by carrying out promotion activities ranging from distribution<br />

<strong>of</strong> brochures to holding health festivals etc.<br />

Clinic Based Health Promotion<br />

These can be carried out at the clinic level, providing individual and group<br />

counseling for promoting smoking cessation and discouraging other risky<br />

behaviors. Health educators and dietitians can have a stronger role in this regard.<br />

Specific Protection<br />

This role is at present limited to the provision <strong>of</strong> vaccination in the clinic for<br />

protection against human papilloma virus, the causative agent for cervical cancer<br />

in women and hepatitis vaccinations?<br />

Secondary Prevention<br />

Early Detection: Screening<br />

Several cancers can be detected early. For many <strong>of</strong> the cancers there is a component<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 85


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

<strong>of</strong> patient education on being made aware <strong>of</strong> the early signs <strong>of</strong> cancer and self<br />

examination. Screening <strong>of</strong> cancers requires physical examination by physician,<br />

while others require a specific procedure such as Pap smear or a diagnostic<br />

laboratory or radiology test. Depending on resources such screening facilities may<br />

or may not be available at the primary care level and may the family doctor may<br />

have to refer a patient to a tertiary care facility to get these done.<br />

Prompt Treatment Referral:<br />

The primary care physician is ideally suited to refer a patient for confirmation<br />

<strong>of</strong> a screening result by additional diagnostic workup and for prompt referral to<br />

access treatment.<br />

High risk Patient Monitoring<br />

Monitoring <strong>of</strong> patients with family history <strong>of</strong> cancer or with other evidence <strong>of</strong><br />

higher risk can be better followed up by the primary care physician, once the<br />

high risk is established.<br />

Tertiary Prevention<br />

Rehabilitation & Palliative care<br />

Having a trusted long term relationship with the patient and his/her family the<br />

primary care physician can be instrumental in the decisions and care process <strong>of</strong><br />

those diagnosed with cancer, particularly in rehabilitation and palliative care.<br />

This can be more effective if the primary care setup is advanced to cover home<br />

health services.<br />

Cancer Specific Roles<br />

This section highlights the role <strong>of</strong> the primary care physician limited to the<br />

activities in the clinic. The additional roles as described above in the general role<br />

section still apply.<br />

The family doctor <strong>of</strong>fice would require some operational changes to be effective<br />

in clinical prevention:<br />

1. Having a database (electronic or register) <strong>of</strong> patients under care and to be<br />

able to contact them by a calling system or an SMS system to come for<br />

preventive checkup is critical.<br />

2. Incorporation <strong>of</strong> information technology can be very useful in alerting<br />

patients and reminding doctors, evenly distributing preventive care over a<br />

period and follow up on results. If the medical record is paper-based then<br />

alerts such as having a checklist, a separator in the file for prevention care<br />

and periodically reviewing preventive care delivered may be sufficient.<br />

3. Cancer preventive care can be initiated by nurses, receptionists, nurse assistants<br />

for all non-high risk patients, so the process <strong>of</strong> ordering some tests can be<br />

initiated by the support staff. It would require some training, to be given by the<br />

busy family doctor. The physician can follow up on the results with the patient.<br />

4. Having a counseling team including a dietitian and health educator is<br />

essential. Physicians at the primary care rarely have time to give good<br />

counseling regarding cancer prevention.<br />

5. Facilities such as having a screening mammogram, or setup for pap smear<br />

collection may vary from practice to practice. Group practices are more able<br />

to provide such services, however nurses can be trained to do pap smears.<br />

The detail <strong>of</strong> the level <strong>of</strong> evidence for a screening procedure, when to carry out<br />

a particular procedure and which patient population is high risk for closer follow<br />

up, are given elsewhere in this manuscript so the emphasis will be on the practical<br />

aspects <strong>of</strong> cancer prevention in the clinical setting.<br />

Colon Cancer<br />

1. Health education with brochures, videos and group sessions either by self or<br />

by the health educator about benefits <strong>of</strong> colon cancer screening, high fiber<br />

diet and low red meat diet.<br />

2. Laboratory in clinic or nurses are to teach patient how to do a heme occult test.<br />

3. Physician to interpret the results and do the flex-sigmoidoscopy if trained and<br />

has the setup or refer the patient to the gastroenterologist for colonoscopy.<br />

A well organized setup may help schedule an appointment and help prepare<br />

patient for colonoscopy.<br />

4. Aspirin prophylaxis to be considered on a case by case basis.<br />

5. High risk patients <strong>of</strong> familial polyposis, inflammatory bowel disease to be<br />

followed as per guidelines.<br />

Lung Cancer<br />

1. Health education with brochures, videos on the hazards <strong>of</strong> smoking and<br />

benefits <strong>of</strong> not smoking and group sessions either by self or by the health<br />

educator are essential. Smoking is a risk factor for multiple other cancers,<br />

but it is specifically mentioned here because <strong>of</strong> being a causative agent.<br />

2. Having a smoking cessation program in the clinic, with group sessions,<br />

counseling, nicotine replacement therapy, antidepressants and or vareniciline<br />

etc should be an essential arsenal <strong>of</strong> every primary care physician.<br />

Skin Cancer<br />

1. Health education with brochures, videos on the hazards <strong>of</strong> excessive UV<br />

light (sunlight) exposure, benefits <strong>of</strong> SPF usage and teaching patients self<br />

examination <strong>of</strong> changes in moles (especially those with higher risk; lighter<br />

skin, multiple moles, elderly (nasolabial folds for basal cell Ca) in group<br />

sessions either by self or by the health educator.<br />

2. Physicians may also do periodic skin examination <strong>of</strong> patients with high risk<br />

for skin Ca, e.g. those with family history, although there is insufficient<br />

evidence for this screening procedure.<br />

Breast Cancer<br />

1. Health education with brochures, videos on breast cancer, benefits <strong>of</strong><br />

screening mammogram, and teaching patients self examination <strong>of</strong> breasts,<br />

in group sessions either by self or by the health educator.<br />

2. Scheduling screening mammogram periodically for all eligible patients with<br />

normal risk and those with high risk. For those not in the high risk category<br />

can be scheduled by the nurse. Patients need to be explained pre-hand about<br />

the slight discomfort <strong>of</strong> the mammography procedure in the primary care<br />

physician <strong>of</strong>fice by the nurse.<br />

3. Physician to break the results to the patient if normal or abnormal and setup<br />

diagnostic mammogram if needed and counsel patient if results positive.<br />

4. High risk determination to be carried out by the physician by screening history<br />

questions exploring family history and advise on genetic marker screening,<br />

followed by mammography on a schedule as per guidelines.<br />

Cervical Cancer<br />

1. Health education with brochures on cervical cancer, benefits <strong>of</strong> pap smear<br />

screening and vaccination along with individual counseling either by self<br />

or by health educator.<br />

2. Have available in clinic anti-HPV vaccine and setup to deliver it by nursing<br />

staff who can prepare patients for it and also educate patients on its possible<br />

side-effects.<br />

3. Having a setup <strong>of</strong> conducting pap smear in clinic, either by self or nurse /<br />

midwife depending upon the size and scope <strong>of</strong> practice with full explanation<br />

86 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


<strong>of</strong> procedure to patient. If such a setup is not possible referring patients<br />

periodically to where the procedure can be carried out.<br />

4. Interpreting results according to protocol and providing patient support in<br />

case <strong>of</strong> positive results.<br />

Thyroid Cancer<br />

1. No recommendation on thyroid cancer but it is in the top-ten list <strong>of</strong> cancers<br />

among Saudi women, some local screening procedure should be considered<br />

for research. Ultrasound examination is fast becoming an extension <strong>of</strong> the<br />

hands <strong>of</strong> the primary care physician worldwide, it may be considered as a<br />

screening tool for research.<br />

2. Health education with brochures on thyroid cancer, instruction on self neck<br />

examination, by self or health educator.<br />

3. Patients with history <strong>of</strong> radiation to neck or family history <strong>of</strong> thyroid cancer<br />

are to be followed closely for periodic examination / ultrasound as per<br />

recommendations.<br />

Uterine Cancer<br />

1. Increase awareness through brochures to peri-menopausal women about<br />

the risks and symptoms <strong>of</strong> uterine cancer along with other elements <strong>of</strong><br />

post-menopausal health.<br />

2. Follow patients with family history <strong>of</strong> uterine cancer more closely as per<br />

guidelines.<br />

Prostate Cancer<br />

1. Educate patients about prostate cancer through brochures and group sessions<br />

by self or by health educator.<br />

2. Consider periodic PSA and DRE for patients in high risk category, such as<br />

those with darker skin African heritage or family history <strong>of</strong> prostate cancer<br />

3. Physicians explain risks and benefits <strong>of</strong> a positive result and guide patients<br />

through the decision making process <strong>of</strong> a positive result.<br />

Oral Cancer<br />

1. Health education on the hazards <strong>of</strong> smoking, tobacco chewing and use <strong>of</strong><br />

pan leaves with betel nuts and tobacco using brochures, waiting room videos<br />

and group sessions by self or health educator.<br />

2. Educate patients on detecting oral painless, precancerous lesions by teaching<br />

them self examination.<br />

3. Physicians or dentists attached to practice to annually examine the oral<br />

cavity <strong>of</strong> high risk patients who smoke or chew tobacco in any form or have<br />

family history <strong>of</strong> oral cancer.<br />

4. Have a smoking cessation / tobacco chewing quitting program in the clinic.<br />

5. Physician to promptly refer patients in case a precancerous lesion or a painless<br />

ulcer is detected by oral cavity examination.<br />

Testicular Cancer<br />

1. Health education brochures on un-descended testes and self-examination <strong>of</strong><br />

testes should be available in the clinic<br />

2. Carryout thorough school physical examination <strong>of</strong> boys to rule out any<br />

un-descended testes.<br />

3. Educate parents <strong>of</strong> boys with un-descended testes on the benefits <strong>of</strong> surgical<br />

correction and hazards <strong>of</strong> un-descended testes (abdominal). Other high<br />

risk cases such as those with family history are to be followed closely with<br />

education on self-examination and physician examination.<br />

Leukemia<br />

1. No recommendation but primary care physician to make a prompt referral to<br />

hematologist on incidental discovery <strong>of</strong> unusually high WBC on CBC done<br />

for some other purpose or suspicion <strong>of</strong> leukemia by any other diagnostic<br />

indicator.<br />

2. Patients with family history are to be followed up as per recommended<br />

guidelines.<br />

Retinoblastoma<br />

1. No recommendation but primary care physicians must be alert on checking<br />

a red reflex in infants and toddlers.<br />

2. Health education material should exist in clinic to alert parents about yellow<br />

/ white reflex in infants among other alerts for this age.<br />

3. Primary care physicians should promptly refer patient to ophthalmologist<br />

for any reflex other than red. .<br />

Future Role<br />

1. Oncogene screening <strong>of</strong> buccal mucosa cells obtained from saliva using<br />

microchips in the primary care clinic may become a reality <strong>of</strong> the near<br />

future, expected to be broadly available within the next 5-10 years, as the<br />

human genome project progresses to detected newer genes. Customized<br />

monitoring and prevention counseling for patients possessing oncogenes is<br />

an immediate outcome with the availability <strong>of</strong> the genetic testing in family<br />

doctor’s <strong>of</strong>fice. Gene therapy may take longer but prevention counseling to<br />

alter modifiable risk factors would become the responsibility <strong>of</strong> the primary<br />

care physician for which preparations are needed to adjust at various levels<br />

in healthcare industry.<br />

2. Use <strong>of</strong> ultrasound in the primary care setup is fast becoming a useful tool.<br />

Its application in cancer screening is researchable particularly for cancers<br />

that are diagnosed late, e.g. pancreatic, ovarian and thyroid etc.<br />

3. Family physician can play a significant role in tumor registry by having a<br />

cancer detection program, and a notifiable disease (cancer) reporting system<br />

for the number screened monthly and detail <strong>of</strong> those found to be positive.<br />

Training <strong>of</strong> Family Physicians<br />

It is vital that primary care physicians and their staff are trained in early detection <strong>of</strong><br />

cancer, through regular programs and are well supported to deliver these services.<br />

Reference<br />

USPSTF Clinical Prevention Guidelines<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 87


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

ROLE OF PRIMARY CARE PHYSICIAN IN THE PREVENTION AND<br />

EARLY DETECTION OF CANCER: AN ONCOLOGIST PERSPECTIVE<br />

Sedky, L.<br />

Salmania <strong>Medical</strong> Complex, Bahrain<br />

Corresponding Author: Pr<strong>of</strong>. Lobna Sedky, MD<br />

Pr<strong>of</strong>essor <strong>of</strong> Clinical <strong>Oncology</strong>, Cairo University<br />

Consultant, <strong>Medical</strong> <strong>Oncology</strong>, Salmania <strong>Medical</strong> Complex, Kingdom <strong>of</strong> Bahrain<br />

E-mail: lo_sedky@yahoo.com<br />

Abstract<br />

Background: It is now recognized that cancer can be preventable, at early<br />

premalignant phases that provide further opportunities for intervention. Primary<br />

Care Physicians (PCP) play a pivotal role in cancer control by identifying those<br />

individuals whose behavior, environment, and/or heredity characteristics place<br />

them at increased risk for developing cancer. They are in a unique position to<br />

provide life care, which includes recognizing the need for and recommending<br />

cancer prevention and early detection when appropriate.<br />

Material and Method: We reviewed the current status <strong>of</strong> primary care cancer<br />

prevention service and science, highlighting barriers against the role <strong>of</strong> PCPs in<br />

prevention and early detection <strong>of</strong> cancer. Furthermore, we explored approaches that<br />

could help primary care program to achieve its full potential in cancer prevention<br />

and screening.<br />

Results: Primary care clinicians face competing demands, conflicting guidelines,<br />

and lack <strong>of</strong> systems that support provision <strong>of</strong> preventive services. The evidencebased<br />

data on what works for behavioral counseling is especially weak, because<br />

actual adoption <strong>of</strong> the guidelines into practice has been slow and inadequate,<br />

analysis <strong>of</strong> available literature reported a compliance rate <strong>of</strong> only 20% to 60%.<br />

Conclusion: The physicians intellectually support the worth <strong>of</strong> primary prevention,<br />

but can not easily insert and/or rationalize it into the culture and expectations <strong>of</strong><br />

clinical practice. Therefore, a persistent public health objective is to develop and<br />

implement strategies to overcome the barriers that deter provision <strong>of</strong> primary<br />

preventive services.<br />

Introduction<br />

Until recently, the practice <strong>of</strong> oncology has focused principally on intervening<br />

to slow or reverse cancer. Insights from molecular biology and molecular and<br />

population epidemiology justify interventions within a broadened definition <strong>of</strong><br />

carcinogenesis that includes the continuum <strong>of</strong> events from the initial genetic or<br />

epigenetic "hit" to the terminal events. It is now recognized that cancer can be a<br />

preventable, late stage <strong>of</strong> the <strong>of</strong>ten lengthy disease continuum <strong>of</strong> carcinogenesis,<br />

which has reversible early, premalignant phases that provide further opportunities<br />

for intervention.<br />

Family physicians can play an invaluable role in caring for patients. Continuity<br />

<strong>of</strong> care and multigenerational relationships allow a family physician to guide a<br />

patient and family through the referral process with a unique knowledge <strong>of</strong> the<br />

patient's values, family issues, and communication style. Because <strong>of</strong> the close<br />

relationship that primary care physicians <strong>of</strong>ten have with their patients, they are<br />

in a unique position to provide life care, which includes recognizing the need for<br />

and recommending cancer prevention and early detection when appropriate. 1<br />

Family physicians play a pivotal role in cancer control by identifying those<br />

individuals whose behavior, environment, and/or heredity characteristics place them<br />

at increased risk for developing cancer.2 The busy primary care physician (PCP)<br />

is <strong>of</strong>ten knowledgeable about prospective medicine including cancer prevention<br />

& early detection, and most feel it is an important part <strong>of</strong> their practice. However,<br />

they find difficulty in integrating the necessary health maintenance measures into<br />

a busy <strong>of</strong>fice practice.<br />

Background<br />

Primary care plays a central role in promoting cancer prevention to the public but<br />

has not achieved its full potential in this regard. We will describe the current status<br />

<strong>of</strong> primary care cancer prevention service and science then explore approaches that<br />

could help primary care achieve its full potential in cancer prevention and screening.<br />

The role <strong>of</strong> health care practitioner includes: Diagnosing, delivering physical care<br />

and treatment, educating patients and family members, assessing psychosocial<br />

strengths and referring for needed services. In addition, <strong>of</strong>fering emotional support,<br />

assisting in maintaining positive outlook, and advocating for best care.3<br />

The current status reveals that most people see their primary care clinician several<br />

times a year and many rely on primary care for screening and behavior advice.<br />

However, when those who are not up-to-date for cancer screening are asked why?<br />

the most common reply is, "my doctor didn't recommend it." Recent studies have<br />

enriched our understanding <strong>of</strong> the processes by which cancer preventive services<br />

are provided in primary care. Primary care clinicians face competing demands,<br />

conflicting guidelines, and lack <strong>of</strong> systems that support provision <strong>of</strong> preventive<br />

services. The evidence-based data on what works for behavioral counseling is<br />

especially weak. Appropriate models and tools to support delivery <strong>of</strong> first-rate<br />

cancer prevention care are being developed. New laboratories for study called<br />

Practice Based Research Networks are in place but widespread adoption lags.<br />

Evidence from high quality resources, such as Put Prevention into Practice,<br />

Cancer Planet, and recommendations <strong>of</strong> the US Preventive Services Task Force,<br />

are readily available but seldom consulted or applied.4<br />

Prevention is defined as the reduction <strong>of</strong> cancer mortality via reduction in the<br />

incidence <strong>of</strong> cancer. This can be accomplished by avoiding a carcinogen or altering<br />

its metabolism, pursuing lifestyle or dietary practices that modify cancer-causing<br />

factors or genetic predispositions; and/or medical intervention (chemoprevention)<br />

to successfully reverse pre-neoplastic changes.5<br />

Much <strong>of</strong> the promise for cancer prevention comes from observational epidemiologic<br />

studies that show associations between modifiable lifestyle factors or environmental<br />

exposures and specific cancers. Evidence is now emerging from randomized<br />

controlled trials designed to test whether interventions suggested by the<br />

epidemiologic studies, as well as leads based on laboratory research, result in<br />

reduced cancer incidence and mortality.6<br />

While physicians are key to primary preventive care, their delivery rate is suboptimal.<br />

The prevailing PCP model was the "one-stop-shop" physician who could<br />

provide anything from primary to tertiary care, but whose provision was dominated<br />

by the delivery <strong>of</strong> immediate diagnoses and treatments, namely secondary care. The<br />

secondary/tertiary prevention PCP model sustained the expectation <strong>of</strong> immediacy<br />

<strong>of</strong> corrective action, cure, and satisfaction sought by patients and physicians<br />

alike, and, thereby, de-prioritized primary prevention in practice in favor <strong>of</strong> the<br />

immediate benefits <strong>of</strong> secondary care.7<br />

Barriers <strong>Against</strong> The Role <strong>of</strong> PCPs in Prevention and Early Detection <strong>of</strong><br />

Cancer<br />

The majority <strong>of</strong> primary care physicians (PCPs) particularly family physicians,<br />

concur with the preventive care guidelines and agree that it is their responsibility<br />

to deliver preventive care services. However, actual adoption <strong>of</strong> the guidelines<br />

into practice has been slow and inadequate, studies report a compliance rate <strong>of</strong><br />

88 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


only 20% to 60%. Consequently, the lack <strong>of</strong> preventive care delivery translates to<br />

lost opportunities to decrease morbidity and mortality via primary and secondary<br />

prevention. Furthermore, spending time to discuss prevention with a patient was<br />

perceived by some physicians as not being a prominent element in the role <strong>of</strong><br />

doctor nor an effective use <strong>of</strong> physician time, hence, the task <strong>of</strong> prevention could<br />

be delegated to other members <strong>of</strong> the medical team.8<br />

In the case <strong>of</strong> preventive services guidelines, implementation needs to go beyond<br />

traditional dissemination and promotion efforts to recognize the added patient and<br />

clinician barriers that affect preventive care.9 These barriers include:<br />

1. Clinicians' ambivalence about whether preventive medicine is part <strong>of</strong> their job<br />

2. The psychological and practical challenges that patients face in changing<br />

behaviors<br />

3. Lack <strong>of</strong> access to health care or <strong>of</strong> insurance coverage for preventive services<br />

<strong>of</strong>fered to some patients.<br />

4. Competing pressures within the context <strong>of</strong> shorter <strong>of</strong>fice visits and lack <strong>of</strong> time<br />

5. The lack <strong>of</strong> organized systems in most practices to ensure the delivery <strong>of</strong><br />

preventive care recommendation.<br />

The physicians intellectually support the worth <strong>of</strong> primary prevention, but can<br />

not easily insert and/or rationalize it into the culture and expectations <strong>of</strong> clinical<br />

practice. Therefore, a persistent public health objective is to develop and implement<br />

strategies to overcome the barriers that deter provision <strong>of</strong> primary preventive<br />

services. A necessary goal <strong>of</strong> such strategies would be to raise the perceived<br />

worth and priority <strong>of</strong> primary prevention within the PCP community, furthermore<br />

physicians need to expand their self-perceived clinical role to take fuller advantage<br />

<strong>of</strong> their unique position to deliver primary preventive care.10<br />

Implementation Strategies<br />

The following practice strategies were recommended to help to overcome these barriers:1<br />

1. Adopt a scientifically based preventive protocol<br />

PCP should be familiar with rational screening criteria,11 together with<br />

recommendations <strong>of</strong> experts and accredited organizations like, Canadian Task<br />

Force,12 American Cancer Society13, US preventive services task force,14,15<br />

and American College <strong>of</strong> Physician.16,17 Meanwhile, they must establish an<br />

evidence-based health-maintenance protocol appropriate to their situation and<br />

benefits, with a strong preventive segment that deal with behavioral modifications<br />

and early diagnosis for the public. This protocol must contain enough flexibility<br />

to accommodate different cohorts <strong>of</strong> patients having a variety <strong>of</strong> risk factors.<br />

Meanwhile, to be applicable to a wide variety <strong>of</strong> health care situations including<br />

public hospitals, clinics, health centers, as well as group and individuals practices.<br />

2. Development <strong>of</strong> a Preventive Attitude<br />

Within the physician who in turn could translate it into educating him or her self<br />

in preventive and screening procedures. The PCP must learn to communicate with<br />

their patients in a positive, enthusiastic way to stimulate them to consider being<br />

subjected to cancer control measures and early detection plan.<br />

3. Engage the Patient<br />

Strategies must be developed to have patients as partners to share the responsibility<br />

for health maintenance. Using patient's handouts, which can stress this responsibility,<br />

and educate them about the preventive protocol. Another tool is the patient's health<br />

diaries which contain health maintenance flow charts to be filled by the patient.<br />

Recently, at higher technical level portable personalized computer smart cards<br />

which are shaped like credit cards, can contain the patient's medical record data.18<br />

4. Institutionalize Prevention & Early Detection<br />

It means committing time and resources to ensure prevention occurs on a regular<br />

basis for all patients. This strategy is the most important and is the one many<br />

practices are reluctant to do. Using clear simple guidelines and accurate easy<br />

methodology for ensuring periodic evaluation feedback. While, it is mandatory<br />

to identify a coordinator and/or auditor responsible for ensuring integrity <strong>of</strong> the<br />

health maintenance tracking system and give feedback about PCP compliance<br />

with the recommended practice.<br />

5. Time Management19<br />

Time saving can be achieved by performing only proven health maintenance<br />

procedures, as well as keeping an organized record system, but these steps alone<br />

may not be enough. The incorporation <strong>of</strong> physician assistants and the use <strong>of</strong><br />

paramedical staff, and nurse practitioners as members <strong>of</strong> the team can be very<br />

helpful.<br />

The Role <strong>of</strong> Cancer Prevention in Practice<br />

Cancers occur not as a sudden catastrophic events, but rather as a the result <strong>of</strong> a<br />

complex and long-evolving process. Carcinogenesis can take decades to evolve<br />

completely, providing time and opportunity to intervene to stop or to reverse its<br />

progress either before the clinical appearance <strong>of</strong> cancer or at its earliest stages. Due<br />

to to the continuing burden, public health interventions have focused on prevention<br />

and early detection to reduce cancer incidence and mortality.20<br />

Behavior change is a difficult task for both patient and PCP. Physicians believe<br />

that implementing patient behavior change required changing the patient's<br />

mindset, yet significant barriers were related to physicians themselves.21 They<br />

acknowledged their lack <strong>of</strong> training, knowledge, and skill in behavior change<br />

process and recommendation conveyance. With participation <strong>of</strong> both patient and<br />

PCP about cancer, in addition to the development <strong>of</strong> a strong preventive attitude,<br />

the stage will be set for the long term appropriate prevention and early detection<br />

implementing strategy. For most individuals who are not symptomatic for cancer<br />

and in good health, unless a physician suggests their participation in a prevention<br />

study, they are likely to remain unaware <strong>of</strong> this option.<br />

As cancer prevention has matured and proved its role in the science and practice <strong>of</strong><br />

oncology. The American Society <strong>of</strong> Clinical <strong>Oncology</strong> (ASCO) has strengthened its<br />

commitment to cancer prevention by establishing its Cancer Prevention Committee<br />

(CAPC) in 2002. With the major objectives are to improve preventive interventions,<br />

expand these efforts globally, also to collaborate with FDA on regulatory issues<br />

involved with preventive drug development.6 Another positive mark, was<br />

publication <strong>of</strong> the comprehensive Institute <strong>of</strong> Medicine report, which <strong>of</strong>fers<br />

recommendations to increase the rates <strong>of</strong> adoption, the reach, and the impact <strong>of</strong><br />

evidence-based cancer prevention and early detection interventions.8<br />

Since cancer can be caused by a variety <strong>of</strong> different factors and may develop over a<br />

number <strong>of</strong> years, therefore some risk factors can be controlled. Choosing the right<br />

health behaviors and preventing exposure to certain environmental risk factors<br />

can help prevent the development <strong>of</strong> cancer. For these reasons, it is important to<br />

follow national trends to monitor the reduction <strong>of</strong> these risk factors which focus<br />

on national trends data from two major groups <strong>of</strong> risk factors: Behavioral and<br />

Environmental factors.22<br />

I. Behavioral Factors<br />

Scientists estimate that as many as 50–75 percent <strong>of</strong> cancer deaths are caused by<br />

human behaviors such as smoking, physical inactivity, and poor dietary choices.<br />

Major reduction in cancer incidence are possible through improved nutrition,<br />

physical activity, and avoidance <strong>of</strong> tobacco products. The latter being the only<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 89


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

strategy with demonstrated efficacy and broad applicability. Behavioral trends<br />

that can help to prevent cancer.<br />

- Tobacco Use:23<br />

The most consistent finding, over decades <strong>of</strong> research is the strong association<br />

between tobacco use and cancers <strong>of</strong> many sites. Hundreds <strong>of</strong> epidemiologic<br />

studies have confirmed this association. Further support comes from the fact that<br />

lung cancer death rates in the United States have mirrored smoking patterns with<br />

increases in smoking followed by dramatic increases in lung cancer death rates,<br />

and more recently decreases in smoking followed by decreases in lung cancer<br />

death rates in men.<br />

While smoking causes about 30% <strong>of</strong> all deaths from cancer. Avoiding both<br />

smoked and smokeless tobacco use is the single most important step people<br />

can take to reduce the cancer burden. Taking into consideration, age at smoking<br />

whether youth or adult smoking, time <strong>of</strong> quitting, health pr<strong>of</strong>essional advice and<br />

recommendations to quit.<br />

- Diet:24<br />

It has been estimated that dietary factors are responsible for at least one third <strong>of</strong><br />

cancer mortality. Meanwhile, there is overwhelming evidence that modifiable<br />

features <strong>of</strong> lifestyle most notably nutrition dominate in the disease.25 As a general<br />

rule, epidemiologic studies have suggested association between diet and cancer<br />

development, but prospective observational or interventional studies have not<br />

provided strong support.<br />

PCPs are potentially the most suitable instrument to implement change in diet<br />

habits as a strategy to prevent cancer. However, several obstacles are encountered,<br />

such as the fact that most physicians are poorly educated in nutrition, also reactive<br />

methods <strong>of</strong> prevention are valued more than proactive modalities. Not surprisingly,<br />

standard medical textbooks commonly acknowledge data linking diet with cancer<br />

yet underestimate the role <strong>of</strong> nutrition in cancer prevention.<br />

Based on population-based epidemiologic data, health care pr<strong>of</strong>essionals should<br />

lead a health advocacy strategy to promote for a healthy weight and encourage<br />

eating a moderate-fat high fiber diet, with enough fruits and vegetables while<br />

limiting consumption <strong>of</strong> red meat which may help to prevent breast and CRC<br />

cancers.26 While avoiding too much alcohol consumption is also an important<br />

step in reducing head & neck cancer risk.27 Multivitamin and mineral supplements<br />

have been advocated for cancer prevention, but the evidence is insufficient.<br />

It is worth noted that children should be targeted in health promotion campaigns,<br />

as studies have shown that counseling parents about nutrition can affect children's<br />

food choices.28<br />

- Physical Activity:29<br />

Obesity and physical inactivity cause about 25–30 % <strong>of</strong> several <strong>of</strong> the major<br />

cancers including colon, breast, endometrial, kidney, and esophageal cancers.30<br />

Obesity is estimated to cause 14 % <strong>of</strong> cancer deaths in men and 20 % <strong>of</strong> cancer<br />

deaths in women.31<br />

Numerous studies have convincingly demonstrated evidence that undertaking<br />

and maintaining moderate levels <strong>of</strong> physical activity rather than hard ones<br />

confers protective effects against cancer. In addition, they provide a rational<br />

for incorporating physical activity counseling as part <strong>of</strong> routine practice in the<br />

primary care setting.<br />

Consensus reports have recommended moderate physical activity 30 min a day on<br />

most days <strong>of</strong> the week (3 hrs/wk ). Recommendations to health care pr<strong>of</strong>essionals32<br />

suggested adoption <strong>of</strong> a physical activity strategy based on an intensive and<br />

sustained exercise counseling by visits, calls, and newsletters. Meanwhile to<br />

ensure effectiveness <strong>of</strong> such a policy in the community, involving schools through<br />

physical education classes, after hours recreation, availability <strong>of</strong> play areas, and<br />

providing healthy snack foods in school settings. Similarly, promote the availability<br />

<strong>of</strong> equipments and trainers for older individuals. Thus, physical activity counseling<br />

should become component <strong>of</strong> routine practice in the primary care setting.<br />

II. Environmental Factors<br />

Data associated with environmental exposures and their relationship to cancer<br />

development are reported, such as second hand tobacco smoke (also known as<br />

environmental tobacco smoke), ionizing radiation, ultraviolet radiation, chemical<br />

exposures as pesticides & toxins, as well as biological agents. Infections may<br />

also be associated with cancer development, HPV infection is a necessary event<br />

for subsequent cancer cervix. Likewise, EBV has been associated with Burkitt’s<br />

lymphoma and Helicobacter pylori with gastric cancer.<br />

Prevention can be accomplished by avoiding a carcinogen, or early detection and<br />

treatment <strong>of</strong> the precancerous lesion through a sustained follow up <strong>of</strong> individuals<br />

known to be at risk. Hence, the role <strong>of</strong> PCP in the primary care health center.33<br />

Cancer Risk Assessment and Screening: Facts and Recommendations<br />

For a growing number <strong>of</strong> preventive services, available data are sufficiently robust<br />

to quantify the magnitude <strong>of</strong> benefits and harms for specific population groups,<br />

but this precision gives rise to difficult ethical questions about trade-<strong>of</strong>fs.34 If a<br />

preventive service poses potential benefits and harms, some would recommend that<br />

avoid making any generic recommendations. Instead uniformly advocate shared<br />

decision making, in which the clinician reviews the trade-<strong>of</strong>fs with patients and<br />

helps them decide for themselves based on personal preferences. This approach,<br />

however, may be impractical and ethically unnecessary except for "close calls" in<br />

which judgments about whether benefits outweigh harms fluctuate dramatically<br />

based on personal preferences. Even in those cases, a large proportion <strong>of</strong> patients<br />

expects the clinician to give advice.35<br />

Cancer risk assessment begins in the primary care clinician’s <strong>of</strong>fice. Essential<br />

components <strong>of</strong> that process include:2<br />

1. Documentation <strong>of</strong> personal and family cancer information.<br />

2. Identification <strong>of</strong> families at increased risk for cancer.<br />

3. Modification <strong>of</strong> cancer screening recommendations according to degree<br />

<strong>of</strong> risk.<br />

4. Referral <strong>of</strong> high-risk individuals to cancer genetics clinics.<br />

The risk assessment criteria would be helpful for physicians such as the one<br />

compiled by Hample et al36 that stratify family history into average, moderate,<br />

and high genetic risk establishes a threshold for referring patients to cancer<br />

genetics clinics. Individuals categorized as average risk should follow general<br />

population guidelines for cancer screening. Moderate risk persons require increased<br />

surveillance <strong>of</strong> at-risk organs, whereas high risk groups require cancer genetics<br />

counseling as well as increased surveillance protocols.37 (Please refer to disease<br />

specific manuscripts in this issue).<br />

Whereby encountered patients whose family histories suggest an increased genetic<br />

risk for cancer without meeting criteria for specific hereditary cancer mutations;<br />

these individuals may benefit from modified cancer screening protocols and<br />

other risk reduction measures. Although most cancer genetic risk manifests in<br />

adulthood, identification <strong>of</strong> families at increased genetic risk for cancer may be<br />

lifesaving even in childhood.38<br />

At all levels <strong>of</strong> cancer risk, families influence adherence to cancer screening<br />

and surveillance recommendations. Individuals who test positive for cancerassociated<br />

mutation may feel less encouraged to express their feeling within<br />

apparent emotions. While, those who test negative may experience survivor guilt.<br />

A review <strong>of</strong> randomized controlled trials involving cancer screening programs<br />

90 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


evealed that providing patients with an individualized risk estimate, increases<br />

the probability that they will participate in these programs.39<br />

Genetic Counseling<br />

For most individuals, a positive family history <strong>of</strong> cancer confers negligible or<br />

only slight additional risk. In some patient, however, the family history suggests<br />

a genetic predisposition to cancer that requires modified screening strategies<br />

compared with the general population. Rarely, the family history is suggestive <strong>of</strong><br />

a hereditary cancer syndrome that warrants referral to a cancer genetics specialist.<br />

With the promulgation <strong>of</strong> guidelines for management <strong>of</strong> persons at increased<br />

genetic risk for cancer and the availability <strong>of</strong> genetic tests to identify those with<br />

hereditary cancer syndromes family physicians play an increasingly crucial role<br />

in cancer risk assessment and management.34<br />

Considerable research effort is now devoted to potential venues for gene therapy<br />

for individuals with genetic mutations or polymorphisms that put them at high<br />

risk <strong>of</strong> cancer. Meanwhile, genetic testing for high-risk individuals with enhanced<br />

surveillance or prophylactic surgery for those who test positive is already available<br />

for certain types <strong>of</strong> cancer including breast and colon cancers.40, 41<br />

Recommendations<br />

Towards achieving the full potential <strong>of</strong> primary care, the jump from the guideline<br />

page to the community practice has been a long overdue. Currently, the evidence<br />

is emerging to assure that primary care achieves its potential in cancer prevention<br />

and early detection. <strong>Medical</strong> groups, health plans, and policy makers will need to<br />

support evidence based change processes for practices, that subsequently lead to<br />

evidence based care processes in the examination room and in the lives <strong>of</strong> patients.<br />

Proposed standard operating procedures (SOPs) guidelines <strong>of</strong> the role <strong>of</strong> PCP in<br />

cancer prevention & early detection:<br />

1. First visit documentation <strong>of</strong> patient's health pr<strong>of</strong>ile to identify the individual<br />

or families at risk for cancer.<br />

2. Proper generation and maintenance <strong>of</strong> patient records, and make sure to be<br />

completed and adequate over time.<br />

3. Annual visit is scheduled on the patient's birthday.<br />

4. PCP must be patient, appreciating <strong>of</strong> patient's fear, communicate in a positive<br />

enthusiastic way.<br />

5. PCP should have enough time to explain and discuss the risk factors<br />

for cancer and the role <strong>of</strong> physical activity, healthy diet, screening tests,<br />

chemoprevention, maintenance <strong>of</strong> health records, and prophylactic surgical<br />

procedures.<br />

6. Schedule the patient at risk for the suitable screening workup<br />

7. Assurance <strong>of</strong> low risk patients<br />

8. Referral <strong>of</strong> high risk cases for genetic counseling<br />

9. Coordinate referral to curative procedures<br />

10. Referral to dietitian when indicated<br />

11. The support staff may include physician assistants, paramedical personnel,<br />

and nurse practitioner. With supply <strong>of</strong> educational opportunities as well as<br />

the time and equipment to address the needs <strong>of</strong> the patient.<br />

12. Physician assistants can provide continuity and <strong>of</strong>fer health maintenance<br />

as part <strong>of</strong> their job,<br />

13. Nurses and clerical <strong>of</strong>fice personnel can play a role in scheduling, referral<br />

and reminder calls.<br />

14. Use <strong>of</strong> handouts, brochures, CDs about the risk factors and preventive<br />

measures<br />

15. Auditing within the team to ensure continuity <strong>of</strong> a high standard program<br />

16. Regular refreshing workshops for the PCP and team to maintain update <strong>of</strong><br />

evidence-based procedures and protocols.<br />

17. To develop continuous medical educational programs (CME) at all levels<br />

to provide for the educational needs <strong>of</strong> the PCPs who will carry the burden<br />

<strong>of</strong> cancer control in the community.<br />

18. Cancer-control measures must be coordinated with other health services as<br />

cardiac, respiratory, metabolic, accidents, and alcohol abuse.<br />

19. Major items <strong>of</strong> cancer risk reduction using changing lifestyle measures<br />

should be part <strong>of</strong> the routine visit to PCP. The health pr<strong>of</strong>essional must<br />

advice the patient verbally, handle flyers, send newsletters, and supply<br />

referral services as part <strong>of</strong> the main cancer control strategy. The healthy<br />

lifestyle recommendations include:<br />

A. Make healthy food choices<br />

a. Eat foods high in fiber - try to increase the amount <strong>of</strong> fiber in your diet<br />

to between 20 and 30 grams daily. High-fiber foods include whole grains,<br />

fruits and vegetables.<br />

b. Limit processed foods, sweets and salt.<br />

c. Avoid foods high in saturated fats.<br />

d. Eat 5 <strong>of</strong> more servings <strong>of</strong> fruits and vegetables daily.<br />

e. Choose foods rich in omega-3 fatty acids.<br />

f. Don’t overeat. Watch portion size and calories.<br />

g. Limit sweets.<br />

B. Avoid alcohol consumption.<br />

C. Maintain a healthy weight.<br />

D. Engage in regular physical activity, preferably 45 to 60 minutes five<br />

days per week.<br />

E. Don’t smoke.<br />

F. Get regular check-ups and talk to your primary care doctor about regular<br />

cancer screenings.<br />

20. Enhance the relationship with policy makers to increase the effectiveness<br />

<strong>of</strong> cancer prevention and control activities nationwide<br />

21. Expand the use <strong>of</strong> information technology in cancer surveillance, particularly<br />

in cancer registries.<br />

22. Conducting research designed to help the cancer community better understand<br />

the factors that increase cancer risk and identify opportunities to prevent<br />

cancer.<br />

Conclusion<br />

It is estimated that 70% or 80% <strong>of</strong> cancer could be controlled if preventive measures<br />

which primarily involve lifestyle modification and early diagnostic procedures<br />

could be instituted in all our population.<br />

Clinical preventive medicine for PCPs means <strong>of</strong>fering a comprehensive preventive<br />

package for each patient. Cancer prevention is an important part <strong>of</strong> this package.<br />

Integrating prevention into the fabric <strong>of</strong> primary care practice is an important<br />

primary care challenge. Nevertheless, cancer prevention is a major part <strong>of</strong> this<br />

effort. PCPs must adopt an evidence based protocol, engage the patient, and<br />

most importantly commit resources to institutionalize clinical cancer prevention<br />

and early detection policy. In the past decade, concepts <strong>of</strong> selective longitudinal<br />

health maintenance have replaced the previous teaching that all adults should have<br />

a complete annual physical checkup.66 It is true that PCPs are advisors, yet they<br />

should act as enforcers as regards the <strong>of</strong> cancer control.<br />

If the patient at risk can be convinced that their lives will be longer, better, and<br />

more productive because <strong>of</strong> prospective health care, at an affordable price, they<br />

will be more likely to seek this kind <strong>of</strong> care and advice from their PCP on a<br />

continuing basis.<br />

The PCP's location in the community places him in a unique position to deliver<br />

the necessary health measures to control cancer within the community. Family<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 91


cancer care in the arab world | march 23-25, 2010 | riyadh, ksa <<br />

physicians are therefore now more than ever apart <strong>of</strong> the multidisciplinary cancer<br />

care team.<br />

Delegating the task <strong>of</strong> primary prevention counseling and education to a team which<br />

beside the PCP includes as well nutritionists, nurse-educators, health-educators,<br />

or other trained medical staff could act as a viable alternative<br />

References<br />

1. Frame PS, and Werth PL. How primary health care providers can integrate<br />

cancer prevention into practice.Cancer 1993;72:1132-7.<br />

2. Tyler CV, & Snyder CW, Cancer risk assessment: Examining the family<br />

physician's role. J Am Board Fam Med 2006: 19; 468-77.<br />

3. David S Rosenthal. Cancer care: The family physician's role in the era <strong>of</strong><br />

improving survivorship. The 50th annual meeting <strong>of</strong> American Academy <strong>of</strong> Family<br />

Physicians. 1998 Medscape portals<br />

4. Woolf SH, and Atkins D. The evolving role <strong>of</strong> prevention in health care.<br />

Contributions <strong>of</strong> the USPS task force. AM J Prev Med 2001; 20 (3s): 13-20<br />

5. Austoker J. Cancer prevention in primary care: Screening for ovarian, prostatic,<br />

and testicular cancers. BMJ 1994; 309: 315-20<br />

6. Lippman SM, Levin B, Brenner DE, et al. cancer prevention and the ASCO. JCO<br />

22: 3848-51, 2004<br />

7. Gramling R, Nash J, Siren K, et al. Family physician self efficacy with screening<br />

for inherited cancer risk. Annals <strong>of</strong> Fam Med ; Aol 2, March/April 2004<br />

8. Ganz PA, Kwan L, Somerfield MR, et al. The role <strong>of</strong> prevention in oncology<br />

practice: Results from a 2004 survey <strong>of</strong> ASCO membes. JCO 24: 2948-57, 2006<br />

9. Frame PS. Health maintenance in clinical practice: Strategies & barriers. Am<br />

Fam Phys 1992; 45: 1192-200<br />

10. NCI, US NIH. Cancer prevention overview health pr<strong>of</strong>essional version, 2007.<br />

11. Frame P.S. Critical review <strong>of</strong> adult health maintenance: part 3: prevention <strong>of</strong><br />

cancer. J Fam Practice 1986; 22: 6: 511-20<br />

12. Canadian Task Force on the Periodic Health Examination. The periodic health<br />

examination. Can Med Assoc J 1979; 121:1194-1254<br />

13. American Cancer Society. Summary <strong>of</strong> current guidelines for the cancerrelated<br />

checkup: recommendations. New York: American Cancer Society: 1988.<br />

14. U.S. Preventive Services Task Force. Guide to clinical preventive services,<br />

2nd ed. Washington, DC: Office <strong>of</strong> Disease Prevention and Health Promotion,<br />

U.S. Government Printing Office, 1996.<br />

15. U.S. Preventive Services Task Force. Put Prevention Into Practice. Clinician's<br />

handbook <strong>of</strong> preventive services, 2nd ed. (Available from the Agency for Healthcare<br />

Research & Quality. Pub. No. APPIP 98-0025.) Washington, DC: Office <strong>of</strong> Disease<br />

Prevention and Health Promotion, 1998.<br />

16. Eddy DM. A manual for assessing health practices and designing practice<br />

policies: the explicit approach. Philadelphia: American College <strong>of</strong> Physicians,<br />

1992.<br />

17. Eddy DM. Common screening tests. Philadelphia: American College <strong>of</strong><br />

Physicians, 1991.<br />

18. Dickey LL, Petitti D. Assessment <strong>of</strong> a patient-held mini-record for adult health<br />

maintenance. J Fam Pract 1990; 31:431-8.<br />

19. Frame PS, Wetterau NW, Paarey B. A model for the use <strong>of</strong> physician's assistants<br />

in primary care. J Fam Pract 1978; 7: 1195-201<br />

20. Leslie G Ford, Lori M Minasian, Worta McCaskill-Stevens, et al. Prevention<br />

and early detection clinical trials: Opportunities for primary care providers and<br />

their patients. Ca Cancer J Clin 2003;53: 82<br />

21. Mirand AL, Beehler GP, Kuo CL, Mahoney MC. Physician perceptions <strong>of</strong><br />

primary prevention: qualitative base for the conceptual shaping <strong>of</strong> a practice<br />

intervention tool. BMC Public Health. 2002; 2:16. 10.1186/ 1471-2458-2-16.<br />

22. U.S. Department <strong>of</strong> Health and Human Services. Healthy People 2010, 2nd<br />

ed. Understanding and improving health and objectives for improving health. 2<br />

vols. Washington, DC: Government Printing Office, Nov 2000.<br />

23. NIH consensus and state <strong>of</strong> the science conference: statement on tobacco use,<br />

prevention, cessation, and control. Vol23, N 3, June 12-14, 2006<br />

24. Norman J Temple. Nutrition in cancer prevention: An integrated approach. J<br />

Am College <strong>of</strong> Nutrition. Vol 21,N2, 79-83, 2002<br />

25. World cancer Research Fund " Food, nutrition and the prevention <strong>of</strong> cancer:<br />

A global perspective" Washington DC Am Inst <strong>of</strong> Ca Res, 1997<br />

26. Prentice RL, Caan B, Chlebowski RT, et al. Low-fat dietary pattern and risk<br />

<strong>of</strong> invasive breast cancer: the Women's Health Initiative Randomized Controlled<br />

Dietary Modification trial. JAMA 2006;295: 629-642<br />

27. Terry MB, Zhang FF, Kabat G, et al. Lifetime alcohol intake and breast cancer<br />

risk. Ann Epidemiol 2006; 16: 230-40<br />

28. Hursti UK: Factors influencing children's food choice. Am Med 31: 26-32, 1999<br />

29. Tehard B, Friedenreich CM, Oppert JM, et al. Effect <strong>of</strong> physical activity on<br />

women at increased risk <strong>of</strong> breast cancer: results from the E3N cohort study.<br />

Cancer Epidemiol Biomarkers Prev. 2006;15:57-64<br />

30. Eliassen AH, Colkditz GA, Rosner B, et al. Adult weight change and risk <strong>of</strong><br />

postmenopausal breast cancer. JAMA 2006;296: 193-201<br />

31. Chakravarthy MV, Joyner MJ, and Booth FW. An obligation for PCP to<br />

prescribe physical activity to sedentary patients to reduce the risk <strong>of</strong> chronic<br />

health conditions. Mayo clinic proc, 2002;77:165-73<br />

32. Lawlor DA, Hanratty B. The effect <strong>of</strong> physical activity advice given in routine<br />

primary care consultations: A systematic review. J Public health med. 2001;23:219-<br />

26<br />

33. Cancer prevention Overview Health Pr<strong>of</strong>essional Version-NCI, modified<br />

07/17/2007<br />

34. Acheson LS, Weiser GL, Zyzanski MA, et al.Family history taking in community<br />

family practice: Implications for genetic screening. Genet Med. 2000;2:180-85<br />

35. Edwards A, Unigwe S, Elwyn G, et al. Effects <strong>of</strong> communicating individual risks<br />

in screening programs: Cochrane systematic review. BMJ 2003; 327:703-9.<br />

36. Hample H, Sweet K, Westman JA, et al. Referral for cancer genetics<br />

consultation: a review and compilation <strong>of</strong> risk assessment criteria. J Med Genet<br />

2004;41:81-91<br />

37. ASCO special article: ASCO policy statement update: genetic testing for cancer<br />

susceptibility. JCO, Vol 21, N 12, June 15, 2003;2397-2406<br />

38. Rose P. Evaluation <strong>of</strong> questionnaire on cancer family history, on general<br />

practice. GPs reassure those at low risk and referre those at high risk. BMJ<br />

2000; 320: 187<br />

39. Edwards A, Unidwe S, Elwyn G, et al. Effects <strong>of</strong> communicating individual<br />

risks in screening programs: Cochrane systematic review. BMJ 2003;327:703-9<br />

40. McInerney-Leo A, Biesecker BB, Hadley DW, et al. BRCA 1/2 testing in<br />

hereditary breast and ovarian cancer families, II: Impact on relationships. Am J<br />

Med Genet A 2005;133:165-9<br />

41. Burt R, Neklason DW. Genetic testing for inherited colon cancer.<br />

Gastroenterology 2005; 128 (6):1696-1716.<br />

42. Gail MH, Costantino JP. Validating and improving models for projecting the<br />

absolute risk <strong>of</strong> breast cancer. J NCI 2001:93:334-35<br />

43. Anderson GL, Chlebowski RT, Rossouw JE, et al. Prior hormone therapy<br />

and breast cancer risk in the WHI randomized trial <strong>of</strong> estrogen plus progestin.<br />

Maturitas 2006; 55:103-105<br />

44. Weiss LK, Burkman RT, Cushing-Haugen KL, et al. HRT regimens and breast<br />

cancer risk. OB/GYN 2002;100(6):1148-58<br />

45. Ault KA: Future II Study Group. Effect <strong>of</strong> prophylactic HPV L1 virus like-particle<br />

vaccine on risk <strong>of</strong> cervical IEN grade 2/3 and adeno-carcinoma in situ: a combined<br />

92 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


analysis <strong>of</strong> four randomized clinical trials. Lancet 2007;369(9576):1861-68<br />

46. NCCN Guidelines, version 2, 2008<br />

47. Winawer SJ, Zauber AG, Fletcher RH, et al. Guidelines for Colonoscopy<br />

Surveillance after Polypectomy: A Concensus Update by the US Multi-Society<br />

Task Force on Colorectal Cancer and the American Cancer Society. CA Cancer<br />

J Clin. 2006;56(3):143-159.<br />

48. Loeb S, Roehl KA, Antenor JA, Catalona WJ, Suarez BK, Nadier RB. Baseline<br />

prostate-specific antigen compared with median prostate-specific antigen for<br />

age group as predictor <strong>of</strong> prostate cancer risk in men younger than 60 years old.<br />

Urology. 2006:67:316-320.<br />

49. Gerald Andriole, et al. Serial Prostate Cancer Screening Findings Reviewed.<br />

the Prostate, Lung, Colorectal and Ovarian (PLCO) Project Team. BJU lnt.<br />

2008;102:1524-1530<br />

50. Babaian RJ, Naya Y, Cheli C, Ha F. The detection and potential economic value<br />

<strong>of</strong> complexed prostate specific antigen and a first line test. J Urol. 2006;175:897-<br />

901.<br />

51. Fisher B, Constantino JP, Wickerham DL, et al. Tamoxifen for prevention <strong>of</strong><br />

breast cancer: report <strong>of</strong> the National Surgical Adjuvant Breast and Bowel Project<br />

P-1 Study. J Natl Cancer Inst. 1998;90:1371-1388.<br />

52. Vogel VG, Costantion JP, Wickerham DL, et al. Effects <strong>of</strong> tamoxifen vs raloxifene<br />

on the risk <strong>of</strong> developing breast cancer and other disease outcomes. The NSABP<br />

Study <strong>of</strong> Tamoxifen & raloxifene (STAR) P-2 trial. JAMA 2006;295:2727-1<br />

53. Cuzick J . Powles T, veronesi U et al. Overview <strong>of</strong> the main outcomes in<br />

breast=cancer prevention trials. Lancet 2003;361- 300<br />

54. Ettinger B, Black DM, Mitlak BH, et al. Reduction <strong>of</strong> vertebral fracture risk<br />

in postmenopausal women with osteoporosis treated with raloxifene: results from<br />

a 3-year randomized clinical trial. Multiple Outcomes <strong>of</strong> Raloxifene Evaluation<br />

(MORE) Investigators. JAMA. 1999;282:637-645.<br />

55. Martino S, Cualey JA, Barrett-Connor E, et al. Continuing outcomes relevant<br />

to Evista: breast cancer incidence in post menopausal osteoporotic women in a<br />

randomized trial <strong>of</strong> raloxifene. J Natl Cancer Inst. 2004;96:1751-1761.<br />

56. Ford JM, Whittemore AS. Predicting and Preventing Hereditary CRC. JAMA<br />

2006; 296: 1521- 23<br />

57. Markowitz LE, Dunne EF, Saraiya M, et al: Centers for Disease Control<br />

and Prevention (CDC): Advisory Committee on Immunization Practices (ACIP).<br />

Quadrivalent Human Papillomavirus Vaccine: Recommendations <strong>of</strong> the Advisory<br />

Committee on Immunization Practices (ACIP). MMWR Recomm Rep 2007;56(RR-<br />

2):1-24.<br />

58. Future II Study Group.Quadrivalent vaccine against HPV to prevent high<br />

grade cervical lesions. N Engl J Med 2007; 356: 1915-26<br />

59. Saslow D, Castle PE, Cox JT, et al. American cancer Society Guideline for<br />

HPV vaccine use to prevent cervical cancer and its precusors. Ca Cancer J Clin<br />

2007; 57(1): 7-28<br />

60. Brawer MK, Lin DW, Williford WO, et al. Effect <strong>of</strong> finasteride and/or terazosin<br />

on serum PSA: results <strong>of</strong> VA Cooperative Study #359. Prostate 1999;39:234-239.<br />

61. Guess HA, Gormley GJ, Stoner E, et al. The effect <strong>of</strong> finasteride on prostate<br />

specific antigen: Review. J Urol 1996; 155: 3-9<br />

62. Task Force on Community Preventive Services. Introducing the Guide to Clinical<br />

Preventive Services: methods, first recommendations and expert commentary. Am<br />

J Prev Med 2000;18(suppl 1):1-142<br />

63. Rebbeck TR, Friebel T, Lynch HT, et al. Bilateral prophylactic mastectomy<br />

reduces breast cancer risk in BRCA1 and BRCA2 mutation carriers: the PROSE<br />

Study Group. J Clin Oncol. 2004; 22:1055-1062.<br />

64. Eisen A, Lubinski J, Klijn J, et al. Breast cancer risk following bilateral<br />

oophorectomy in BRCA1 and BRCA2 mutation carriers: an international case-<br />

control study. J Clin Oncol. 2005;23:7491-7496.<br />

65. UK testicular cancer study group. Etiology <strong>of</strong> testicular cancer association<br />

with congenital abnormalities, age at puberty, infertility, and exercise. BMJ<br />

1994; 308: 1393 – 9<br />

66. Han P.K. Historical changes in the objectives <strong>of</strong> the periodic health examination.<br />

Ann Intern Med 1997;127:910-917.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 93


news from the arab world <<br />

SAVE THE DATE!<br />

ASCO
Multidisciplinary
Cancer
Management
Course
<br />

(MCMC)

<br />

Hosted
by:
Gulf
International
Cancer
Center
<br />

Abu
Dhabi,
UAE.
<br />

February 25 – 27, 2010<br />

President <strong>of</strong> the Conference International ASCO Course Director<br />

Pr<strong>of</strong>. Aly Abdel Razek Pr<strong>of</strong>. Hugo Villar<br />


<br />



<br />

ASCO:
American
Society
<strong>of</strong>
Clinical
<strong>Oncology</strong>


<br />

SEMCO:
South
&
East
Mediterranean
College
<strong>of</strong>
<br />

<strong>Oncology</strong>


<br />


<br />


<br />

For
more
information,
please
contact
Pr<strong>of</strong>.
Dr.
Ahmed
Elzawawy

<br />

E‐mail:

worldcooperation@gmail.com
OR
ahmedelzawawy@hotmail.com<br />

94 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


H E V E N T O G R O U P<br />

SAVE THE DATE!<br />

www.winteracademy.net<br />

ST. MORITZ AND PONTRESINA – APRIL 8-11, 2010<br />

KEY TOPICS<br />

Breast cancer: adjuvant therapy<br />

Colorectal cancer<br />

Gastrointestinal – non colon: update<br />

Gynecological session<br />

Non small cell lung cancer (NLSC)<br />

Genitourinary cancer<br />

Focus on new drugs<br />

Anti-angiogenic agents:<br />

from biology to clinical trials<br />

Chemotherapy side effects<br />

PRESIDENTS<br />

Pr<strong>of</strong>. Francesco Cognetti, Italy<br />

Pr<strong>of</strong>. Francesco Di Costanzo, Italy<br />

Pr<strong>of</strong>. Sami Khatib, Jordan<br />

INTERNATIONAL BOARD<br />

Co-Chairman Hamdy Abdel Azim, Egypt<br />

Omalkhair Abulkhair, KSA<br />

Said Al Natour, Jordan<br />

Assem Al Radi, KSA<br />

Aguilar Enrique Aranda, Spain<br />

Sandro Barni, Italy<br />

Corrado Boni, Italy<br />

Adda Bounedjar, Algeria<br />

Eric Van Cutsem, Belgium<br />

Mircea Dediu, Romania<br />

Hussein Hadi Hashmi, Libya<br />

Alan Horwich, UK<br />

Joseph Kattan, Lebanon<br />

Gurunath Kilara, India<br />

Claus-Henning Köhne, Germany<br />

Paris A. Kosmidis, Greece<br />

Stefan Madajewicz, USA<br />

Mervat Saud Al-Saleh, Kuwait<br />

Mohsen Mokhtar, Egypt<br />

Giovanni Rosti, Italy<br />

Armando Santoro, Italy<br />

Michael Untch, Germany<br />

Yasser Abd El Kader, Egypt<br />

Shahinaz Bedri, Sudan<br />

Fawaz Deirawan, Syria<br />

Khaled Ahmad Al Saleh, Kuwait<br />

First EURO-ARAB Congress.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 95<br />

2 ND EDITION


news from the arab world <<br />

Confirmed Invited speakers:<br />

Raul Ribeiro (USA) Maarten Egeler (Netherlands)<br />

Sima Jeha (USA) Stefan J. Friedrichsdorf (USA)<br />

Monica Metzger (USA) Don Aaronson (Netherlands)<br />

Sheri Spunt (USA) Mohab Ayas (KSA)<br />

Matt Krasin (USA) Miguel Abboud (Lebanon)<br />

Eric Bouffet (Canada) Sara Day (USA)<br />

Victor Blanchette (Canada) Huda Huijer (Lebanon)<br />

96 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 97


news from the arab world <<br />

Conference on Topics in Therapeutic and Diagnostic<br />

<strong>Medical</strong> Physics<br />

Amman, Jordan<br />

Dead Sea Resort<br />

April 28th – May 2 nd , 2010<br />

Sponsored By:<br />

The American <strong>Association</strong> <strong>of</strong> Physicists in Medicine<br />

(AAPM)<br />

International Scientific Exchange Program (ISEP)<br />

Third <strong>Arab</strong> Radiology Conference (ARC)<br />

<strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer (AMAAC)<br />

Jordan <strong>Medical</strong> Physics Society (JMPS)<br />

Program Director<br />

Dr. Adel Mustafa, AAPM, USA<br />

Co-Directors<br />

Dr. Sami Al-Khatib, Secretary General, AMAAC, Jordan<br />

Dr. Hazem Haboub, Chair <strong>of</strong> ARC Committee, Jordan<br />

Dr. Mustafa Al-Majali, President <strong>of</strong> JMPS, Jordan<br />

Dr. Samir Khraisat, President <strong>of</strong> Jordan Radiology Society, Jordan<br />

Associate Directors<br />

Dr. Maysoon Al-Taher, AMAAC, Jordan<br />

Dr. Matthew Al-Ghazi, AAPM, USA<br />

For more details please contact Dr. Maysoon Al-Taher at<br />

maysoontahir@hotmail.com<br />

98 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 < 99


news from the arab world <<br />

Amman - Jordan<br />

July 29-31, 2010<br />

Conference<br />

Topics:<br />

• Breast Tumors • GI/GIST Tumors<br />

• Lymphoma/Leukemia • Lung Tumors<br />

• Palliative Care & <strong>Oncology</strong> Nursing<br />

Abstracts:<br />

• Awards will be granted to best 3 abstracts<br />

• Deadline for submitting abstracts is May 31, 2010<br />

For more details, you can contact Dr. Jamal Khader at:<br />

Jkhader@khcc.jo<br />

100 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


notes <<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10< 101


cancer awareness calendar <<br />

january<br />

february<br />

march<br />

april<br />

may<br />

june<br />

july<br />

august<br />

september<br />

october<br />

november<br />

december<br />

Cervical Cancer Awareness Month<br />

Screening and Early Detection Awareness Month<br />

Colorectal Cancer Awareness Month<br />

Cancer Fatigue Awareness Month<br />

Melanoma and Skin Cancer Awareness Month<br />

National Cancer Survivors Day<br />

Sarcoma Awareness Month<br />

Pain Medicine and Palliative Care<br />

Gynecologic Cancer Awareness Month<br />

Prostate Cancer Awareness Month<br />

Leukemia and Lymphoma Awareness Month<br />

Breast Cancer Awareness Month<br />

Lung Cancer Awareness Month<br />

Smoking Cessation<br />

5 A Day Awareness Month<br />

102 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


objectives & scope <strong>of</strong> the PAJO <<br />

The <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> (PAJO) is the <strong>of</strong>ficial <strong>Journal</strong> <strong>of</strong> the <strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer (AMAAC). It is a<br />

quarterly publication targeting health pr<strong>of</strong>essionals interested in the oncology field. It is a multidisciplinary peer-reviewed journal that<br />

publishes articles addressing medical oncology, malignant hematology, surgery, radiotherapy, pediatric oncology, geriatric oncology,<br />

basic research and the comprehensive management <strong>of</strong> patients with malignant diseases in addition to international oncology activities,<br />

congresses & news.<br />

The journal will be addressed, as a first step, mainly to the pr<strong>of</strong>essionals in the hematology & oncology field in the Middle East region<br />

and North Africa. The goal is to share local & regional research activities news and to be updated with international activities.<br />

We hope, with your support, to achieve our following objectives:<br />

1. Promote and encourage research activities in the <strong>Arab</strong> World.<br />

2. Disseminate & analyze epidemiological local, regional and international data.<br />

3. Update health pr<strong>of</strong>essionals with the most recent advances, news & developments in the field <strong>of</strong> oncology.<br />

4. Improve the level <strong>of</strong> scientific publications arising form the <strong>Arab</strong> World.<br />

5. Keep health pr<strong>of</strong>essionals connected and exposed to the activities <strong>of</strong> different <strong>Arab</strong> cancer societies.<br />

6. Share with our immigrant compatriots their activities & feedback in this field.<br />

7. Involve all health pr<strong>of</strong>essionals interested in the field <strong>of</strong> <strong>Oncology</strong> within the multidisciplinary scope <strong>of</strong> the <strong>Journal</strong>.<br />

8. Encourage post graduates students to submit their research work.<br />

instructions for authors <<br />

1. Manuscript Categories<br />

1.1. Clinical trials<br />

The Editor-in-Chief and an Associate Editor generally review<br />

Reports from clinical trials. Selected manuscripts are also reviewed<br />

by at least two external peer reviewers. Comments <strong>of</strong>fered by<br />

reviewers are returned to the author(s) for consideration.<br />

Manuscript acceptance is based on many factors, including the<br />

importance <strong>of</strong> the research to the field <strong>of</strong> oncology & the quality<br />

<strong>of</strong> the study. Authors should focus on accuracy, clarity, and brevity<br />

in their presentation, and should avoid lengthy introductions,<br />

repetition <strong>of</strong> data from tables and figures in the text, and unfocused<br />

discussions. Extended patient demographic data should be included<br />

in a table, not listed within the text.<br />

Reports from Clinical trials are limited to 3,000 words <strong>of</strong> body<br />

text, excluding the abstract, references, figures, and tables. They<br />

are limited to six total figures and tables. All abstracts are strictly<br />

limited to 250 words. Titles are to be descriptive, but succinct.<br />

Results <strong>of</strong> clinical studies should be supported by a clear description<br />

<strong>of</strong> the study design, conduct, and analysis methods used to obtain<br />

the results.<br />

Reports <strong>of</strong> phase II & III studies should include from the protocol<br />

a clear definition <strong>of</strong> the primary end point, the hypothesized value<br />

<strong>of</strong> the primary end point that justified the planned sample size,<br />

and a discussion <strong>of</strong> possible weaknesses, such as comparison to<br />

historical controls.<br />

Phase I studies will be well received if they have interesting clinical<br />

responses, unusual toxicity that pointed to mechanism <strong>of</strong> action <strong>of</strong><br />

the agents, and important or novel correlative laboratory studies<br />

associated with the trials.<br />

1.2. Review Articles<br />

All reviews must be clinically oriented, ie, at least half the review<br />

must describe studies that detail human impact, marker effect on<br />

prognosis, or clinical trials.<br />

Review Articles should be prepared in accordance with the <strong>Journal</strong>’s<br />

Manuscript Preparation Guidelines, and will be reviewed in the<br />

same manner as Reports from Clinical Trials. Reviews are limited<br />

to 4,500 words <strong>of</strong> body text, excluding the abstract, references,<br />

figures, and tables. The editors also suggest a limit <strong>of</strong> 150 references.<br />

1.3. Editorials / Comments / Controversies<br />

The Editor-in-Chief may solicit an Editorial to accompany an<br />

accepted manuscript. Authors who wish to submit unsolicited<br />

Comments and Controversies should contact the Editor-in-Chief,<br />

before submission to determine the appropriateness <strong>of</strong> the topic<br />

for publication in the <strong>Journal</strong>.<br />

Editorials should be no more than four to five pages in length.<br />

1.4. Articles on Health Economics<br />

Articles about health economics (cost <strong>of</strong> disease, cost-effectiveness<br />

<strong>of</strong> drugs, etc) are highly encouraged.<br />

1.5. Case Reports / Correspondence / Special Articles<br />

Correspondence (letters to the Editor) may be in response to a<br />

published article, or a short, free-standing piece expressing an<br />

opinion, describing a unique case, or reporting an observation that<br />

would not qualify as an Original Report. If the Correspondence is<br />

in response to a published article, the Editor-in-Chief may choose<br />

to invite the article’s authors to write a Correspondence reply.<br />

Correspondence should be no longer than three pages in length.<br />

Special Articles present reports, news from international, regional<br />

societies as well as news from our compatriots.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10< 103


instructions for the authors <<br />

2. Manuscript submission procedure<br />

All manuscripts should be submitted in word and PDF format<br />

directly to the Editor-in-Chief by email at the following email:<br />

editorinchief.pajo@yahoo.com.<br />

The manuscript should adhere to the journal requirements. Upon<br />

manuscript submission, corresponding authors must provide<br />

unique e-mail addresses for all contributing authors. Receipt <strong>of</strong><br />

manuscripts will be acknowledged via e-mail. Upon completion <strong>of</strong><br />

editorial review, the corresponding author will receive notification<br />

<strong>of</strong> the Editor’s decision, along with the reviewers’ comments, as<br />

appropriate, via e-mail.<br />

3. Disclosures <strong>of</strong> Potential Conflicts <strong>of</strong> interest<br />

In compliance with standards established and implemented by<br />

ASCO’s Conflict <strong>of</strong> Interest Policy (J Clin Oncol 24:519–521,<br />

2006), it is the PAJO’s intent, as previously referred, to ensure<br />

balance, independence, objectivity, and scientific rigor in all <strong>of</strong> its<br />

editorial policies related to the <strong>Journal</strong> through the disclosure <strong>of</strong><br />

financial interests, among other measures. All contributors to the<br />

<strong>Journal</strong> are required to disclose financial and other relationships<br />

with entities that have investment, licensing, or other commercial<br />

interests in the subject matter under consideration in their<br />

article. These disclosures should include, but are not limited to,<br />

relationships with pharmaceutical and biotechnology companies,<br />

device manufacturers, or other corporations whose products or<br />

services are related to the subject matter <strong>of</strong> the submission.<br />

Disclosures <strong>of</strong> financial interests or relationships involving the<br />

authors must be addressed on the Author Disclosure Declaration<br />

form. The corresponding author may complete the form on behalf<br />

<strong>of</strong> other authors, or authors may complete their own forms and<br />

forward them to the corresponding author. This information will<br />

be sent to the Editorial Board. Statements regarding financial<br />

support <strong>of</strong> the research must be made on the manuscript title page,<br />

and disclosed on the form. This form is available upon request<br />

from the Editorial Office. All disclosures will appear in print at<br />

the end <strong>of</strong> all published articles.<br />

The <strong>Journal</strong> requires all Editors and reviewers to make similar<br />

disclosures. Reviewers are asked to make disclosures when<br />

accepting a review.<br />

4. Manuscript Preparation Guidelines<br />

Title Page<br />

The first page <strong>of</strong> the manuscript must contain the following<br />

information: (1) title <strong>of</strong> the report, as succinct as possible; (2)<br />

author list <strong>of</strong> no more than 20 names (first name, last name); (3)<br />

names <strong>of</strong> the authors’ institutions and an indication <strong>of</strong> each author’s<br />

affiliation; (4) acknowledgments <strong>of</strong> research support; (5) name,<br />

address, telephone and fax numbers, and e-mail address <strong>of</strong> the<br />

corresponding author; (6) running head <strong>of</strong> no more than 80 characters<br />

(including spaces); (7) list <strong>of</strong> where and when the study has been<br />

presented in part elsewhere, if applicable; and (8) disclaimers, if any.<br />

Abstract<br />

Abstracts are limited to 250 words and must appear after the title<br />

page. Abstracts must be formatted according to the following<br />

headings: (1) Purpose, (2) Patients and methods (or materials and<br />

methods, similar heading), (3) Results, and (4) Conclusion. Authors<br />

may use design instead <strong>of</strong> Patients and methods in abstracts <strong>of</strong><br />

Review Articles. Comments and Controversies, Editorials and<br />

Correspondence do not require abstracts.<br />

Text<br />

The body <strong>of</strong> the manuscript should be written as concisely as<br />

possible and must not exceed the manuscript category word<br />

limits described herein. All pages <strong>of</strong> a submission should be<br />

numbered and double-spaced. Helvetica and Arial at 12pt size<br />

are the recommended fonts for all text (see Figures section for<br />

acceptable fonts for figures). The <strong>Journal</strong> adheres to the style<br />

guidelines set forth by the International Committee <strong>of</strong> <strong>Medical</strong><br />

<strong>Journal</strong> Editors.<br />

References<br />

References must be listed and numbered after the body text in the<br />

order in which they are cited in the text. They should be doublespaced<br />

and should appear under the heading “REFERENCES.”<br />

Abbreviations <strong>of</strong> medical periodicals should conform to those<br />

used in the latest edition <strong>of</strong> Index Medicus and on MEDLINE.<br />

The «List <strong>of</strong> <strong>Journal</strong>s Indexed in Index Medicus» includes the<br />

latest abbreviations. Inclusive page numbers must be cited in<br />

the reference. When a reference is for an abstract or supplement,<br />

it must be identified as such in parentheses at the end <strong>of</strong> the<br />

reference. Abstract and supplement numbers should be provided,<br />

if applicable. When a reference is a personal communication,<br />

unpublished data, a manuscript in preparation, or a manuscript<br />

submitted but not in press, it should be included in parentheses in<br />

the body <strong>of</strong> the text, and not cited in the reference list. Published<br />

manuscripts and manuscripts that have been accepted and are<br />

pending publication should be cited in the reference list.<br />

Reference Style<br />

º <strong>Journal</strong> article with one, two, or three authors<br />

1. Dolan ME, Pegg AE: O6-Benzylguanine and its role in<br />

chemotherapy. Clin Cancer Res 8:837-847, 1997<br />

º <strong>Journal</strong> article with more than three authors<br />

2. Knox S, Hoppe RT, Maloney D, et al: Treatment <strong>of</strong> cutaneous<br />

T-cell lymphoma with chimeric anti-CD4 monoclonal antibody.<br />

Blood 87:893-899, 1996<br />

º <strong>Journal</strong> article in press (manuscript has been accepted for<br />

publication)<br />

3. Scadden DT, Schenkein DP, Bernstein Z, et al: Combined<br />

immunotoxin and chemotherapy for AIDS-related non-Hodgkin’s<br />

lymphoma. Cancer (in press)<br />

º Supplement<br />

4. Brusamolino E, Orlandi E, Morra E, et al: Analysis <strong>of</strong> long-term<br />

104 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info


esults and prognostic factors among 138 patients with advanced<br />

Hodgkin’s disease treated with the alternating MOPP/ABVD<br />

chemotherapy. Ann Oncol 5:S53-S57, 1994 (suppl 2)<br />

º Book with a single author<br />

5. Woodruff R: Symptom Control in Advanced Cancer. Victoria,<br />

Australia, Asperula Pty Ltd, 1997, pp 65-69<br />

º Book with multiple authors<br />

6. Iverson C, Flanagin A, Fontanarosa PB, et al: American <strong>Medical</strong><br />

<strong>Association</strong> Manual <strong>of</strong> Style (ed 9). Baltimore, MD, Williams &<br />

Wilkins, 1998<br />

º Chapter in a multiauthored book with editors<br />

7. Seykora JT, Elder DE: Common acquired nevi and dysplastic nevi<br />

as precursor lesions and risk markers <strong>of</strong> melanoma, in Kirkwood<br />

JM (ed): Molecular Diagnosis and Treatment <strong>of</strong> Melanoma. New<br />

York, NY, Marcel Dekker, 1998, pp 55-86<br />

º Abstract<br />

8. Bardia A, Wang AH, Hartmann LC, et al: Physical activity and<br />

risk <strong>of</strong> postmenopausal breast cancer defined by hormone receptor<br />

status and histology: A large prospective cohort study with 18 years<br />

<strong>of</strong> follow up. J Clin Oncol 24:49s, 2006 (suppl; abstr 1002)<br />

9. Kaplan EH, Jones CM, Berger MS: A phase II, open-label,<br />

multicenter study <strong>of</strong> GW572016 in patients with trastuzumab<br />

refractory metastatic breast cancer. Proc Am Soc Clin Oncol<br />

22:245, 2003 (abstr 981)<br />

º Conference/meeting presentation<br />

10. Dupont E, Riviere M, Latreille J, et al: Neovastat: An<br />

inhibitor <strong>of</strong> angiogenesis with anti-cancer activity. Presented at<br />

the American <strong>Association</strong> <strong>of</strong> Cancer Research Special Conference<br />

on Angiogenesis and Cancer, Orlando, FL, January 24-28, 1998<br />

º Internet resource<br />

11. Health Care Financing Administration: Bureau <strong>of</strong> data<br />

management and strategy from the 100% MEDPAR inpatient<br />

hospital fiscal year 1994: All inpatients by diagnosis related groups,<br />

6/95 update. http://www.hcfa.gov/a1194drg.txt<br />

º Digital Object Identifier (DOI)<br />

12. Small EJ, Smith MR, Seaman JJ, et al: Combined analysis <strong>of</strong> two<br />

multicenter, randomized, placebo-controlled studies <strong>of</strong> pamidronate<br />

disodium for the palliation <strong>of</strong> bone pain in men with metastatic<br />

prostate cancer. J Clin Oncol 10.1200/JCO.2003.05.147<br />

º Government Announcement/Publication<br />

13. Miller BA, Ries CAG, Hankey BF, et al (eds): Cancer Statistics<br />

Review: 1973-1989. Bethesda, MD, National Cancer Institute,<br />

NIH publication No. 92-2789, 1992<br />

º ASCO Educational Book<br />

14. Benson AB 3rd: Present and future role <strong>of</strong> prognostic and<br />

predictive markers for patients with colorectal cancer. Am Soc<br />

Clin Oncol Ed Book 187-190, 2006<br />

Figures<br />

Figures must be cited in the order they appear in the text using<br />

<strong>Arab</strong>ic numerals. Figures should be submitted in a seperate<br />

documen. Figure legends are required for all article types. Figure<br />

legends must not exceed 55 words per figure and should be written<br />

below the figure.<br />

Images may be embedded in word or Power Point files.<br />

Tables<br />

Tables must be cited in the order in which they appear in the<br />

text using <strong>Arab</strong>ic numerals. The table’s legend may include any<br />

pertinent notes and must include definitions <strong>of</strong> all abbreviations<br />

and acronyms that have been used in the table. Tables submitted<br />

with multiple parts will be renumbered. Tables should be submitted<br />

in a seperate document. Legends must not exceed 55 words per<br />

table and should be written above the figure.<br />

Appendices/Acknowledgments<br />

Appendices and acknowledgments will appear in the print version<br />

<strong>of</strong> the article.<br />

Language: Appropriate use <strong>of</strong> the English language is encouraged<br />

for publication in the <strong>Journal</strong>.<br />

5. Post-acceptance Information<br />

Copyright Form<br />

Corresponding authors must provide unique e-mail address for each<br />

contributing author at manuscript submission. Upon acceptance <strong>of</strong><br />

the manuscript, each author will receive an e-mail invitation to sign<br />

a statement confirming that the manuscript contains no material for<br />

which publication would violate any copyright or other personal<br />

or proprietary right <strong>of</strong> any person or entity. Manuscripts will not<br />

be published until each author has completed the form.<br />

Page Pro<strong>of</strong>s<br />

Corresponding author will receive pro<strong>of</strong>s and must carefully<br />

review them for data and typesetting errors. Corrections to pro<strong>of</strong>s<br />

must be returned by e-mail, fax, or mail within 1 week. The<br />

corresponding author is responsible for collecting and submitting<br />

all author corrections into a single submission. Publication may<br />

be delayed if pro<strong>of</strong>s are not returned by the publisher’s deadline.<br />

The Editor-in-Chief must approve all major alterations, which<br />

may delay publication <strong>of</strong> the manuscript.<br />

www.amaac.info <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10< 105


subscription to PAJO <<br />

subscribe to PAJO<br />

In case you are not receiving the PAJO or you would like to<br />

change your address, kindly to contact us by email to:<br />

editorinchief.pajo@yahoo.com or by fax to: + 962 65 62 38 53<br />

and update us with the following information:<br />

Title<br />

First Name<br />

Family Name<br />

Country<br />

City<br />

Clinic or Hospital or Office’s Name<br />

Street<br />

Bldg<br />

Floor<br />

Land Phone<br />

Mobile Phone<br />

Fax<br />

E-mail<br />

106 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 3; issue 1 | March 10 www.amaac.info

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!