30.12.2012 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

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<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.org | vol 4; issue 1 | March 2011<br />

Original Articles<br />

• Pathologist’s role in Modern <strong>Oncology</strong><br />

Practice<br />

• Chroidal Metastases from Breast<br />

carcinoma<br />

No winter lasts forever; no spring skips its turn.<br />

~Hal Borland<br />

• Hyp<strong>of</strong>ractionated vs. Conventional<br />

RT in Glioblastoma Multiforme


Aranesp ® (darbepoetin alfa) SureClick Brief Prescribing Information. Please refer to the<br />

Summary <strong>of</strong> Product Characteristics before prescribing Aranesp ® . Pharmaceutical Form:<br />

Solution for injection presented in prefilled pens containing 150, 300, and 500 micrograms <strong>of</strong><br />

darbepoetin alfa, for single-dose use only. Indication: Treatment <strong>of</strong> symptomatic anaemia in adult<br />

cancer patients with non-myeloid malignancies receiving chemotherapy. Dosage and<br />

Administration: Cancer patients: Aranesp ® should be administered by the subcutaneous route to<br />

patients with anaemia (e.g. haemoglobin concentration ≤10 g/dL (6.2 mmol/l)) in order to increase<br />

haemoglobin to not greater than 12 g/dL (7.5 mmol/l). Anaemia symptoms and sequelae may vary<br />

with age, gender, and overall burden <strong>of</strong> disease; a physician’s evaluation <strong>of</strong> the individual patient’s<br />

clinical course and condition is necessary. Due to intra-patient variability, occasional individual<br />

haemoglobin values for a patient above and below the desired haemoglobin level may be<br />

observed. Haemoglobin variability should be addressed through dose management, with<br />

consideration for the haemoglobin target range <strong>of</strong> 10 g/dL (6.2 mmol/l) to 12 g/dL (7.5 mmol/l). A<br />

sustained haemoglobin level <strong>of</strong> greater than 12 g/dL (7.5 mmol/l) should be avoided; guidance for<br />

appropriate dose adjustments for when haemoglobin values exceeding 12 g/dL (7.5 mmol/l) are<br />

observed are described below. The recommended initial dose is 500 μg (6.75 μg/kg) given once<br />

every three weeks, or once weekly dosing can be given at 2.25 μg/kg body weight. If the clinical<br />

response <strong>of</strong> the patient (fatigue, haemoglobin response) is inadequate after nine weeks, further<br />

therapy may not be effective. Aranesp ® therapy should be discontinued approximately four weeks<br />

after the end <strong>of</strong> chemotherapy. Once the therapeutic objective for an individual patient has been<br />

achieved, the dose should be reduced by 25 to 50% in order to ensure that the lowest approved<br />

dose <strong>of</strong> Aranesp ® is used to maintain haemoglobin at a level that controls the symptoms <strong>of</strong><br />

anaemia. Appropriate dose titration between 500 μg, 300 μg, and 150 μg should be considered.<br />

Patients should be monitored closely, if the haemoglobin exceeds 12 g/dL (7.5 mmol/l), the dose<br />

should be reduced by approximately 25 to 50%. Treatment with Aranesp ® should be temporarily<br />

discontinued if haemoglobin levels exceed 13 g/dL (8.1 mmol/l). Therapy should be reinitiated at<br />

approximately 25% lower than the previous dose after haemoglobin levels fall to 12 g/dL (7.5<br />

mmol/l) or below. If the rise in haemoglobin is greater than 2 g/dL (1.25 mmol/l) in 4 weeks, the<br />

dose should be reduced by 25 to 50%. Contraindications: Hypersensitivity to darbepoetin alfa,<br />

r-HuEPO or any <strong>of</strong> the excipients. Poorly controlled hypertension. Special Warnings and<br />

Precautions: General: blood pressure should be monitored in all patients, particularly during<br />

initiation <strong>of</strong> Aranesp ® therapy. If blood pressure is difficult to control by initiation <strong>of</strong> appropriate<br />

measures, the haemoglobin may be reduced by decreasing or withholding the dose <strong>of</strong> Aranesp ® .<br />

Iron status should be evaluated for all patients prior to and during treatment and supplementary<br />

iron therapy may be necessary. Non-response to therapy with Aranesp ® should prompt a search<br />

for causative factors. Deficiencies <strong>of</strong> iron, folic acid or vitamin B12 reduce the effectiveness <strong>of</strong><br />

erythropoiesis-stimulating agents and should therefore be corrected. Intercurrent infections,<br />

inflammatory or traumatic episodes, occult blood loss, haemolysis, severe aluminium toxicity,<br />

underlying haematologic diseases, or bone marrow fibrosis may also compromise the<br />

erythropoietic response. A reticulocyte count should be considered as part <strong>of</strong> the evaluation. If<br />

typical causes <strong>of</strong> non-response are excluded, and the patient has reticulocytopenia, an<br />

examination <strong>of</strong> the bone marrow should be considered. If the bone marrow is consistent with<br />

PRCA, testing for anti-erythropoietin antibodies should be performed. Pure red cell aplasia caused<br />

by neutralising anti-erythropoietin antibodies has been reported in association with erythropoiesisstimulating<br />

agents (ESAs), including darbepoetin alfa. This has been predominantly reported in<br />

Give them the energy<br />

to keep up with life<br />

Aranesp ® <strong>of</strong>fers convenient and tailored treatment<br />

as the only ESA licensed for QW and Q3W 1-3<br />

patients with CRF treated subcutaneously. Cases have also been reported in patients with hepatitis<br />

C treated with interferon and ribavirin, when epoetins are used concomitantly (ESAs are not<br />

indicated for use in this patient population). These antibodies have been shown to cross-react with<br />

all erythropoietic proteins, and patients suspected or confirmed to have neutralising antibodies to<br />

erythropoietin should not be switched to darbepoetin alfa. Active liver disease was an exclusion<br />

criteria in all studies <strong>of</strong> Aranesp ® , therefore no data are available from patients with impaired liver<br />

function. Since the liver is thought to be the principal route <strong>of</strong> elimination <strong>of</strong> Aranesp ® and<br />

r-HuEPO, Aranesp ® should be used with caution in patients with liver disease. Aranesp ® should<br />

also be used with caution in those patients with sickle cell anaemia or epilepsy. Convulsions have<br />

been reported in patients receiving Aranesp ® . Misuse <strong>of</strong> Aranesp ® by healthy persons may lead to<br />

an excessive increase in packed cell volume. This may be associated with life-threatening<br />

complications <strong>of</strong> the cardiovascular system. The needle cover <strong>of</strong> the pre-filled syringe contains dry<br />

natural rubber (a derivative <strong>of</strong> latex), which may cause allergic reactions. In patients with chronic<br />

renal failure, maintenance haemoglobin concentration should not exceed the upper limit <strong>of</strong> the<br />

target haemoglobin concentration. In clinical studies, an increased risk <strong>of</strong> death, serious<br />

cardiovascular events and vascular access thrombosis was observed when ESAs were<br />

administered to target a haemoglobin <strong>of</strong> greater than 12 g/dL (7.5 mmol/l). Controlled clinical trials<br />

have not shown significant benefits attributable to the administration <strong>of</strong> epoetins when<br />

haemoglobin concentration is increased beyond the level necessary to control symptoms <strong>of</strong><br />

anaemia and to avoid blood transfusion. Cancer patients: Effect on tumour growth. Epoetins are<br />

growth factors that primarily stimulate red blood cell production. Erythropoietin receptors may be<br />

expressed on the surface <strong>of</strong> a variety <strong>of</strong> tumour cells. As with all growth factors, there is a concern<br />

that epoetins could stimulate the growth <strong>of</strong> tumours. In several controlled studies, epoetins have<br />

not been shown to improve overall survival or decrease the risk <strong>of</strong> tumour progression in patients<br />

with anaemia associated with cancer. In controlled clinical studies, use <strong>of</strong> Aranesp ® and other<br />

ESAs have shown: shortened time to tumour progression in patients with advanced head and neck<br />

cancer receiving radiation therapy when administered to target a haemoglobin <strong>of</strong> greater than 14<br />

g/dL (8.7 mmol/l) (ESAs are not indicated for use in this patient population); shortened overall<br />

survival and increased deaths attributed to disease progression at 4 months in patients with<br />

metastatic breast cancer receiving chemotherapy when administered to target a haemoglobin <strong>of</strong><br />

12-14 g/dL (7.5-8.7 mmol/l); increased risk <strong>of</strong> death when administered to target a haemoglobin<br />

<strong>of</strong> 12 g/dL (7.5 mmol/l) in patients with active malignant disease receiving neither chemotherapy<br />

nor radiation therapy (ESAs are not indicated for use in this patient population). In view <strong>of</strong> the<br />

above, in some clinical situations blood transfusion should be the preferred treatment for the<br />

management <strong>of</strong> anaemia in patients with cancer. The decision to administer recombinant<br />

erythropoietins should be based on a benefit-risk assessment with the participation <strong>of</strong> the<br />

individual patient, which should take into account the specific clinical context. Factors that should<br />

be considered in this assessment should include the type <strong>of</strong> tumour and its stage; the degree <strong>of</strong><br />

anaemia; life-expectancy; the environment in which the patient is being treated; and patient<br />

preference. In patients with solid tumours or lymphoproliferative malignancies, if the haemoglobin<br />

value exceeds 12 g/dL (7.5 mmol/l), the dosage adaptation described in the Posology and Method<br />

<strong>of</strong> Administration section should be closely respected, in order to minimise the potential risk <strong>of</strong><br />

thromboembolic events. Platelet counts and haemoglobin level should also be monitored at regular<br />

intervals. Pregnancy and Lactation: No adequate experience in human pregnancy and lactation.<br />

Exercise caution when prescribing Aranesp ® to pregnant women. Do not administer to women who<br />

are breastfeeding. When Aranesp ® therapy is absolutely indicated, breastfeeding must be<br />

discontinued. Undesirable Effects: General: There have been reports <strong>of</strong> serious allergic reactions<br />

including anaphylactic reaction, angioedema, allergic bronchospasm, skin rash and urticaria<br />

associated with darbepoetin alfa. Clinical Trial Experience - Cancer patients: Adverse reactions<br />

were determined based on pooled data from seven randomised, double-blind, placebo-controlled<br />

studies <strong>of</strong> Aranesp ® with a total <strong>of</strong> 2112 patients (Aranesp ® 1200, placebo 912). Patients with<br />

solid tumours (e.g., lung, breast, colon, ovarian cancers) and lymphoid malignancies (e.g.,<br />

lymphoma, multiple myeloma) were enrolled in the clinical studies. Incidence <strong>of</strong> undesirable<br />

effects considered related to treatment with Aranesp ® from controlled clinical studies: Very<br />

common (≥1/10) Oedema; Common (≥1/100 to


editorial board < contents <<br />

º Editor-in-Chief<br />

Marwan Ghosn, MD, MHHM<br />

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

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

Lebanon<br />

º Deputy Editor<br />

Sami Khatib, MD<br />

> amaac.pajo@gmail.com<br />

Jordan<br />

º 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 />

Mohamad Jaloudi, MD<br />

> mjaloudi@tawamhospital.ae<br />

UAE<br />

º Design & Layout<br />

Zéna Khairallah<br />

> design@zenak.me<br />

º 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 Sabe’ 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 />

Special Thanks > 4 - 5<br />

Original Articles<br />

º Choroidal Metastases from Breast Carcinoma: Case series<br />

Mohammed Jaloudi et al. > 6 - 10<br />

º Multidrug Resistance- related Protein (MRP) and Lung<br />

Resistance Protein (LRP) mRNA Expression in Egyptian Patients<br />

with Acute Leukemia<br />

Amira Ahmed Hammam et al. > 12 - 16<br />

º Phase II pilot study <strong>of</strong> weekly Docetaxel as Neoadjuvant<br />

Chemotherapy for operable Breast Cancer<br />

Mahmoud Ellithy, et al. > 18 - 23<br />

º The Role <strong>of</strong> Pathologist in Modern <strong>Oncology</strong> Practice<br />

Abdul-Rahman Jazieh et al. > 24 - 26<br />

º Taxanes Versus Vinorelbine and 5-Fluorourocil as a First<br />

Therapy in Metastatic Breast Cancer<br />

Wessam Ali El-Sherief et al. > 28 - 33<br />

º Dose-Dense Chemotherapy in High-Risk Breast Cancer:<br />

Treatment Outcome and Toxicity<br />

Amr El-Kashif et al. > 34 - 39<br />

º Genomics views: Xenobiotic metabolizing enzymes<br />

and cancer risks<br />

Bechr Hamrita et al. > 40 - 46<br />

º Hyp<strong>of</strong>ractionated radiotherapy versus conventional radiotherapy<br />

in treatment <strong>of</strong> glioblastoma multiforme<br />

Mohamed Abdelgawad et al. > 48 - 52<br />

News from the <strong>Arab</strong> World > 54 - 61<br />

º Announcements<br />

º 11th <strong>Pan</strong>arab Cancer Congress<br />

º 1st Annual Congress <strong>of</strong> the <strong>Arab</strong> Alliance for <strong>Medical</strong> Education<br />

º UAE Cancer Congress 2011<br />

º Hematology and Blood Transfusion<br />

º 3rd EASO Masterclass in Clinical <strong>Oncology</strong><br />

º AORTIC 2011<br />

º 3rd Asian Breast Cancer Congress<br />

Cancer Awareness Calendar > 62<br />

Instructions for Authors > 63 - 66<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 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 Mon’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 />

Mauritania Jiddou Abdou, MD<br />

El Issawi Salem Sidi Mohamed, 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 />

UAE Mohammed Jaloudi, MD<br />

Mohamad Abbas Alali, 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 4; issue 1 | March 2011 www.amaac.org


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.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 3


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

Original Article<br />

Special Report<br />

Breast Cancer in Tunisia<br />

Highlights on the Speech and Language<br />

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

Review<br />

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

BLOM Beirut Marathon 08<br />

new publication<br />

<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 2; issue 3 | December 09<br />

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

Original Articles<br />

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

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

breast cancer biomarkers.<br />

radical or palliative basis.<br />

new publication<br />

<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 3 | October 10<br />

Hope begins in the dark, the stubborn hope that if you just show<br />

up and try to do the right thing, the dawn will come. You wait and<br />

watch and work: You don’t give up.<br />

Anne Lamott<br />

Original Articles<br />

• Infiltrating Ductal Carcinomas <strong>of</strong> the • Locally Advanced HNC: Cisplatin and<br />

Breast: Proteomic Analysis <strong>of</strong> Human Docetaxel plus Radiation Therapy<br />

plasma protein<br />

• Triple negative MBC: BRCA1 and EGFR<br />

• Gastric carcinoma: DCF vs ECF<br />

as prognostic biomarkers<br />

<strong>Pan</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 />

Health Economics Review Articles<br />

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

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

advanced NSCLC<br />

advanced prostate cancer<br />

4 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org<br />

new publication<br />

<strong>Pan</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 2; issue 2 | April 09<br />

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

9 TH PAN ARAB CANCER CONGRESS<br />

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

new publication<br />

<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<br />

<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.org | vol 3; issue 4 | December 10<br />

Screening and Early Detection Awareness<br />

Original Articles<br />

• Report <strong>of</strong> preliminary experience <strong>of</strong> the • C/EBPα Expression in Egyptian patients with<br />

prone table stereotactic breast core biopsy Acute Myeloid Leukemia<br />

at the King Fahad National Guard Hospital • Treatment results <strong>of</strong> Stereotactic Radiosurgery<br />

for cerebral arteriovenous malformations<br />

<strong>Pan</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 3 | September 08<br />

Review<br />

Meeting Highlights<br />

Treatment <strong>of</strong> Acute Lymphoblastic Leukemia ASCO 2008<br />

UICC 2008<br />

Targeted Therapy Development<br />

Angiogenesis review<br />

new publication<br />

<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 2; issue 3 | September 09<br />

Original Articles<br />

Low dose Gemcitabine and Cisplatin<br />

in Advanced NSCLC<br />

PRAME and WT1 Genes expression<br />

in CML Patients<br />

While there’s<br />

there’s<br />

life,<br />

hope.<br />

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

Meeting Highlights<br />

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

Best <strong>of</strong> ASCO 2009<br />

new publication<br />

<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 2 | June 10<br />

CANCER<br />

SURVIVOR<br />

MONTH<br />

Original Articles<br />

• Receptor for hyaluronic acid-mediated<br />

• L’ approche immuno-ptoteomique motility(RHAMM/CD168)inAMLpatients<br />

SEPRA et Cancer du Sein (in French)<br />

• Egyptian experience <strong>of</strong> modified<br />

medical thoracoscopy<br />

<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.org | vol 4; issue 1 | March 2011<br />

Original Articles<br />

• Pathologist’s role in Modern <strong>Oncology</strong><br />

Practice<br />

• Chroidal Metastases from Breast<br />

carcinoma<br />

No winter lasts forever; no spring skips its turn.<br />

~Hal Borland<br />

• Hyp<strong>of</strong>ractionated vs. Conventional<br />

RT in Glioblastoma Multiforme


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 />

Géraldine Dalmasso, MD<br />

Kamal El-Dein Hamed Mohamed, MD<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 />

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<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 5


original article <<br />

Choroidal Metastases from Breast Carcinoma: Case series<br />

Jaloudi Mohammad, MD 1 , Sivatrakasam Karthiga, MD 2 , Al Qawasmeh Khaled, MD 1 , Kanbar Jihad, MD 1 , Kumar Pawan, MD 1<br />

(1) Department <strong>of</strong> <strong>Oncology</strong>, Tawam Hospital, Al-Ain, Abu Dhabi, UAE<br />

(2) Department <strong>of</strong> Radiotherapy, Tawam Hospital, Al-Ain, Abu Dhabi, UAE<br />

Corresponding Author: Dr. Mohammed Jaloudi, MD<br />

Department <strong>of</strong> <strong>Oncology</strong>, Tawam Hospital, Al-Ain, Abu Dhabi, UAE<br />

E-mail: mjaloudi@tawamhospital.ae<br />

Key words: Breast Cancer, Ocular, Metastases, Choroids, A-scan & B-scan Ultrasonography, Radiation Therapy.<br />

ISSN: 2070-254X<br />

Introduction<br />

Choroidal metastases were first reported in 1872 by Perls1 , who noted an<br />

intraocular metastatic lung carcinoma. Since then, choroidal metastases have<br />

become well established as the most common intraocular malignancy2 . Choroidal<br />

metastases have not been documented to be a frequent finding in disseminated<br />

breast cancer. Ocular symptomatology in a breast cancer patient should alert the<br />

physician to undertake a thorough examination <strong>of</strong> the eye.<br />

Background<br />

Breast cancer is one <strong>of</strong> the most common malignant diseases with a lifetime<br />

risk <strong>of</strong> approximately 10% in females and is also one <strong>of</strong> the major causes <strong>of</strong><br />

cancer related mortality among women worldwide. Attempts to improve survival<br />

have targeted early diagnosis and development <strong>of</strong> adjuvant and neoadjuvant<br />

chemotherapy regimens. Breast cancer screening programs for early diagnosis<br />

have been incorporated in many international guidelines in the hopes <strong>of</strong><br />

improving survival rates3 .<br />

Frequent or well described sites <strong>of</strong> metastases include bone, liver, lung, pleura,<br />

skin, brain and eye, but tumor manifestations have been reported in nearly<br />

all anatomic regions. Breast cancer possesses the highest incidence <strong>of</strong> ocular<br />

metastatic involvement and the choroids because <strong>of</strong> their high vascularity are<br />

the most common site for ocular metastases. This is followed by the anterior<br />

segment and the optic nerve among solid tumors (female patients with breast<br />

cancer and male patients with lung cancer). The metastatic involvement <strong>of</strong><br />

ocular structures in breast cancer seems to be a rare clinical entity; nevertheless,<br />

histopathological inquiries propose that 10-37% <strong>of</strong> patients with breast cancer<br />

have detectable ocular or orbital metastases2 .<br />

In 12-31% <strong>of</strong> affected individuals, eye metastases were reported to be the<br />

first sign <strong>of</strong> malignant disease or metastatic spread2. Choroidal metastases in<br />

advanced asymptomatic breast cancer patients were determined to be 5% <strong>of</strong><br />

ophthalmological screening. Risk factors included spread <strong>of</strong> disease to more than<br />

one organ and the coexistence <strong>of</strong> lung and brain metastases increased the risk to<br />

11% 4 . The median period from diagnosis <strong>of</strong> breast cancer to the development<br />

<strong>of</strong> choroidal metastases is reported to be 4 years with a median survival <strong>of</strong> 5<br />

to 17 months5 . A large survey <strong>of</strong> 520 eyes with uveal tract metastases revealed<br />

that 88% were located in the choroid, 9% in the iris, and 2% in the ciliary body.<br />

Due to recent advances in breast cancer treatment which has impacted on<br />

improved survival rates, clinicians should begin incorporating a complete<br />

ophthalmological examination for tumoral spread as part <strong>of</strong> a routine screening<br />

program, particularly in high risk patients or in the setting <strong>of</strong> disseminated<br />

disease. This thorough incorporation <strong>of</strong> an ophthalmologic exam and the<br />

improved life expectancy <strong>of</strong> patients with metastatic disease will make the<br />

diagnosis even more prevalent.<br />

Case reports<br />

Six cases <strong>of</strong> Choroidal metastases from breast cancer were detected at Tawam<br />

Hospital, Al-Ain, U.A.E., over a period <strong>of</strong> five years (2004-2008).<br />

These case reports are discussed in brief below.<br />

Case 1… A 50 y/o female with no known co-morbid conditions initially<br />

diagnosed with left breast carcinoma by FNA (fine needle aspiration). Later on<br />

she had a lumpectomy with axillary clearance followed by adjuvant radiotherapy<br />

(T1N0M0) in 1992. Histopathology revealed an infiltrating ductal carcinoma;<br />

grade II, ER/PR positive, Cerb2 negative. All lymph nodes were negative.<br />

She had no further hormonal therapy or chemotherapy. In February 2004, she<br />

had mediastinal lymphadenopathy and lung secondaries. Biopsy confirmed<br />

metastatic IDC, ER positive, PR negative and Cerb2+3. She received 2 cycles<br />

<strong>of</strong> chemotherapy with Docetaxel and Capecitabine. Due to poor tolerance, this<br />

was switched to Docetaxel and Gemcitabine. She completed six cycles with<br />

good response on CT scans. After 8 months she presented with progressive<br />

lymphadenopathy. She was again started on systemic chemotherapy with FEC<br />

(FEC = 5-FU, Epirubicin, Cyclophosphamide). After 4 days in the 2nd cycle,<br />

she began complaining <strong>of</strong> left eye pain and blurring <strong>of</strong> vision. Funduscopic<br />

evaluation by an ophthalmologist revealed whitish sub retinal exudates. Fundus<br />

fluorescein angiography (FFA) and B-scan were consistent with choroidal<br />

metastases. MRI brain and orbits showed normal orbits with small variable<br />

sized lesion in the left frontal, left post parietal, and right high parietal regions,<br />

suggestive <strong>of</strong> cerebral deposits.<br />

She commenced on whole brain radiotherapy including orbits after appropriate<br />

6 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


simulation using 6MV photon beams on LA on 11/07/05. A dose <strong>of</strong> 30 Gy in 10<br />

fractions was delivered using opposing lateral techniques, which was completed<br />

on 24/07/05. After radiotherapy completion; patient lost follow up.<br />

Case 2… A 49 y/o female with no known co morbid conditions was diagnosed<br />

with right breast infiltrating ductal carcinoma with FNA. This was followed by<br />

a right breast lumpectomy with axillary clearance. Histopathology revealed an<br />

infiltrating ductal carcinoma; grade III, Triple negative on 02/06/08. She received<br />

1st cycle <strong>of</strong> chemotherapy; FEC regimen. Post chemotherapy, she was admitted<br />

for febrile neutropenia. During the hospitalization, she began complaining <strong>of</strong> left<br />

eye diplopia. A CT scan <strong>of</strong> the orbits and brain was performed which revealed<br />

a normal brain with left sided retinal detachment, choroidal metastases and optic<br />

nerve involvement.<br />

The patient was evaluated by ophthalmology. On examination, right eye was<br />

normal, left eye disc margins were not clear, yellowish white mass over and<br />

around the disc and inferior part <strong>of</strong> retina. B-scan revealed retinal detachment<br />

with choroidal infiltration. She received radiotherapy to the left orbit at a dose <strong>of</strong><br />

30 Gy in 10 fractions from 31/05/06 to 06/06/06; after radiotherapy completion;<br />

patient lost follow up.<br />

Case 3… An 80 y/o female patient with hypertension, acute renal failure, and<br />

hypercalcaemia was diagnosed with right breast carcinoma via core biopsy in<br />

April 2008. Histopathology was consistent with an infiltrating ductal carcinoma,<br />

ER positive, PR negative, and Cerb2 negative. A bone scan revealed extensive<br />

bone metastases. She was started on the aromatase inhibitor Letrozole. A few<br />

months later, she developed diplopia. A CT scan <strong>of</strong> the brain and orbits revealed a<br />

normal brain with a right orbital mass about 17.0x15.0x7.0 mm along the medial<br />

wall and causing compression <strong>of</strong> the optic nerve. The patient was evaluated<br />

by ophthalmology and later referred to radiotherapy. She received 20 Gy in 5<br />

fractions with conformal technique. Shortly thereafter, she was lost to follow-up.<br />

Case 4… A 49 y/o female with primary infertility was diagnosed with a<br />

triple negative infiltrating ductal carcinoma <strong>of</strong> the right breast on 27/05/2008.<br />

Metastatic workup revealed diffuse bone metastases, including the lumbar<br />

spine. She received 20 Gy in 5 fractions to the lumbar spine. She was then<br />

started on systemic chemotherapy, FEC regimen on 13/06/08. One week later,<br />

she presented with blurred vision in the left eye. An MRI <strong>of</strong> orbits confirmed<br />

choroidal metastases. Upon these findings, she received 30 Gy in 10 fractions<br />

to both orbits including optic nerves from 30/06/08 – 16/07/08. During<br />

radiotherapy, she developed a massive pulmonary embolism which was treated<br />

with anticoagulant. Upon stabilization, she was started on Epirubicin and<br />

Taxotere from 23/07/08 and completed a total <strong>of</strong> 6 cycles with a good PR.<br />

Three months later, she presented with new liver lesions and was started on<br />

Capecitabine and Bevacizumab. Up to last follow-up, she had received 4 cycles<br />

with improvement in her liver lesions.<br />

Case 5… A 62 y/o female was diagnosed with infiltrating ductal carcinoma <strong>of</strong><br />

left breast in June 2005. The tumor was pT3 N2 M0, ER+, PR+, and HER-<br />

2 +. She received adjuvant chemotherapy with 4 FEC and 4 Taxotere with<br />

Trastuzumab. The patient also received radiotherapy to the chest wall and axilla<br />

and maintenance Trastuzumab. Hormonal therapy with an aromatase inhibitor<br />

was started after completion <strong>of</strong> the radiotherapy. In July 2006, she was admitted<br />

for loss <strong>of</strong> vision in the left eye and blurred vision in the right eye. An MRI<br />

revealed bilateral choroidal metastases. She received 40 Gy in 20 fractions to<br />

both eyes between July and August 2006. Hormonal therapy was changed to<br />

Tamoxifen. After radiotherapy, she was started on Capecitabine and Trastuzumab<br />

was recommenced. Unfortunately, she developed a right pathological hip<br />

fracture and expired after the first cycle due to deterioration <strong>of</strong> her condition.<br />

Case 6…. A 38 years old female was diagnosed with left breast cancer in August<br />

2002. She underwent a lumpectomy with axillary clearance. Pathology revealed<br />

invasive lobular carcinoma, grade 2, ER +, PR +, HER-2 positive. Adjuvant<br />

chemotherapy was given with 4 cycles <strong>of</strong> Vinorelbine and Epirubicin (part <strong>of</strong><br />

a clinical trial) followed by 4 cycles <strong>of</strong> Paclitaxel. Adjuvant radiotherapy was<br />

given to the left breast and supra clavicular region for a total <strong>of</strong> 50 Gy in 25<br />

fractions; this was followed by an additional boost to the tumor bed giving 9<br />

Gy in 3 fractions making the total dose <strong>of</strong> radiation therapy given 59 Gy in 28<br />

fractions. Post radiotherapy, she was commenced on Tamoxifen. In September<br />

2004; biopsy proven lung metastases was present. She was restarted on<br />

Docetaxel and Trastuzumab. After 4 cycles and a good PR, a CT scan <strong>of</strong> the chest<br />

revealed progressive disease, so she was switched to Letrozole with Goserelin<br />

and Trastuzumab was continued. In February 2006, a CT scan revealed<br />

progression <strong>of</strong> the lung lesions, Trastuzumab was stopped and patient was<br />

started on Capecitabine and Bevacizumab. From February 2006 until May 2010<br />

patient had several lines <strong>of</strong> chemotherapy. In May 2010 patient presented with<br />

blurred vision in the left eye. MRI <strong>of</strong> the orbits revealed choroidal metastasis<br />

to the left eye. Radiotherapy was given for 25 Gy in 5 fractions to the left eye.<br />

In August 2010 patient was restarted on Trastuzumab, Lapatinib, and Caelyx.<br />

After 4 cycles <strong>of</strong> chemotherapy a CT scan to the chest and abdomen revealed<br />

a new metastatic lesion in the liver. In August 2010 patient was switched to<br />

Ixabepilone, Capecitabine, Lapatinib, and Trastuzumab. Another CT scan in<br />

December 2010 revealed stable disease.<br />

Pathophysiology<br />

Intraocular metastases arise when the neoplasm at the primary site spreads<br />

through the blood to the eye. Due to the high vascularity <strong>of</strong> the uveal and,<br />

specifically, the choroid - there is a greater likelihood that metastases will enter<br />

and remain there. Most choroidal metastases are carcinomas; they are seldom<br />

melanomas or sarcomas6 .<br />

Workup<br />

Workups for choroidal metastases include an MRI <strong>of</strong> the head to rule out brain<br />

metastases and to determine the extent <strong>of</strong> the tumor. Incidence <strong>of</strong> central nervous<br />

system (CNS) metastases increases from 6% to 28% after development <strong>of</strong> ocular<br />

metastases7 . Patients with an unknown primary site <strong>of</strong> origin should obtain a<br />

thorough physical examination and have a chest radiograph and mammogram.<br />

If the primary site is known, serum chemical analysis, plasma carcinoembryonic<br />

antigen (CEA), liver function tests, chest radiography, isotope bone scan, and CT<br />

scan <strong>of</strong> abdomen should be obtained8 .<br />

Treatment<br />

There are a wide variety <strong>of</strong> treatment options for patients with Choroidal<br />

metastases, the choice <strong>of</strong> which depends on many factors, including the<br />

patient’s systemic state, the presence <strong>of</strong> visual symptoms, tumor activity, tumor<br />

size, location, primary site, and whether the patient is concurrently receiving<br />

chemotherapy. The report by Shields et al. presented a breakdown <strong>of</strong> common<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 7


original article <<br />

treatment options and the percentage <strong>of</strong> patients who were receiving these<br />

corresponding therapies. They included: chemotherapy (8%), external beam<br />

radiation (39%), plaque radiation (7%), hormone therapy (1 %), resection (6%),<br />

observation (14%), and combination therapy (24%) 9 .<br />

In cases <strong>of</strong> regressed lesions or visually asymptomatic patients with advanced<br />

systemic disease, the judicious option is to observe6. For visually asymptomatic<br />

patients, many lesions will respond to systemic chemotherapy, hormone<br />

therapy, or immunotherapy. Brinkley10 used a five drug chemotherapy protocol<br />

for treatment <strong>of</strong> metastatic breast cancer and found that choroidal metastases<br />

responded to chemotherapy with the same sensitivity as metastases elsewhere<br />

in body11 . If the lesion is unresponsive to chemotherapy, the patient is visually<br />

symptomatic, or the patient is not able to tolerate chemotherapy, palliative<br />

radiation therapy may be implemented. There are two types <strong>of</strong> radiation:<br />

external beam radiation therapy (teletherapy) and episcleral plaque therapy<br />

(Brachytherapy). Both aim for tumor flattening and pigment proliferation. The<br />

decision on which option to choose depends on the size <strong>of</strong> the lesion and the<br />

patient’s life expectancy.<br />

Because choroidal metastases are radiosensitive, external beam radiation therapy<br />

(EBRT) is the most commonly used treatment for these lesions. This treatment is<br />

administered with the goal <strong>of</strong> saving the vision and the globe and provides quick<br />

tumor regression. The response rate is 80% but if untreated leads to blindness15 .<br />

In some studies chemotherapy was also as effective as radiotherapy16 . The<br />

procedure is performed in an outpatient setting. Radiation from a remote source<br />

is directed at the lesion, and is <strong>of</strong>ten performed with a block to limit the radiation<br />

to anterior ocular structures and to the contra lateral eye. Total doses <strong>of</strong> 30 to 40<br />

Gy are administered in daily doses <strong>of</strong> 2 Gy for 4 weeks.<br />

A study by Rudoler et al <strong>of</strong> 233 eyes with Choroidal metastases treated with EBRT<br />

found that EBRT led to tumor control in 90% <strong>of</strong> eyes, and globe preservation in<br />

98% <strong>of</strong> eyes. Weigel et found that the standardized treatment <strong>of</strong> 40 Gy (with<br />

daily doses <strong>of</strong> 2 Gy given 5 days a week) led to complete remission <strong>of</strong> the tumor<br />

in 38% <strong>of</strong> the patients, partial remission is 27%, minor remission in 18%, and no<br />

remission in 17%.Patients with breast cancer had a significantly increased rate<br />

<strong>of</strong> complete tumor remission, and all patients with combined chemotherapy and<br />

EBRT had complete remission.<br />

Higher dosages produce better tumor regression; however, the risk <strong>of</strong> radiation<br />

retinopathy or radiation- induced ocular complications is greater. Radiation can<br />

cause dry eye, cataract, hair loss, cutaneous erythema, and conjunctival irritation6 .<br />

Late side effects include cataract, glaucoma, and radiation retinopathy12 . Weigel<br />

found that using more than 40 Gy increased the risk <strong>of</strong> radiation retinopathy.<br />

In a retrospective study by Rosset et a112 <strong>of</strong> 58 patients treated with EBRT,<br />

patients treated with higher amounts <strong>of</strong> radiation had better resolution and<br />

acuities, but more complications. Of patients treated with more than 35.5 Gy,<br />

72% had complete resolution (CR) and 18% had partial resolution (PR). Of<br />

patients treated with less than 35.5 Gy, 33% had CR and 41% had PR12. Four<br />

<strong>of</strong> the five patients with complications were found in the group receiving more<br />

than 35.5 Gy.<br />

Episcleral plaque therapy is focal radiotherapy to the eye performed in an<br />

inpatient setting. The benefit is that treatment time is reduced to 3 days. This<br />

therapy is recommended for patients with solitary lesions, patients for whom<br />

other modes <strong>of</strong> treatment have been unsuccessful, or for patients whose life<br />

expectancy is short and who want to maximize their quality <strong>of</strong> life6 . Radioactive<br />

seeds such as Iodine-125 sit inside a gold shield, which is sutured to the globe so<br />

that there is a short distance to the target tissue. The plaque is surgically attached<br />

to the globe at the site <strong>of</strong> the lesion. A typical amount <strong>of</strong> 40 Gy <strong>of</strong> radiation is<br />

delivered to the apex <strong>of</strong> the tumor. Shields15 reported that most patients achieve<br />

tumor regression to half the original thickness and resorption <strong>of</strong> the sub retinal<br />

fluid within the first 3 months after treatment. Complications with plaque therapy<br />

parallel those <strong>of</strong> external beam radiation6 .<br />

Other techniques that are used include local resection, laser photocoagulation,<br />

transpupillary thermotherapy, and enucleation. Transpupillary thermotherapy is<br />

still in the early stages <strong>of</strong> its use in the realm <strong>of</strong> treating choroidal metastases;<br />

however, case reports in the literature have shown promising results13 . When the<br />

patient is in a great deal <strong>of</strong> pain or the choroidal metastases have grown out <strong>of</strong> the<br />

scope <strong>of</strong> treatment options, enucleation is the recommended treatment. Of note,<br />

a recent case report <strong>of</strong> a carcinoid tumor metastatic to the choroid treated with<br />

photodynamic therapy (PDT) showed tumor regression and an improvement in<br />

visual acuity14 .<br />

Survival time<br />

Prognosis is poor for patients diagnosed with choroidal metastases. Average<br />

survival time is 8 to 9 months after diagnosis6. The major determinant <strong>of</strong><br />

prognosis for patients with uveal metastases is based on the primary tumor type.<br />

Cutaneous tumors have the worst prognosis (1 to 2 months survival time), and<br />

breast tumors have the best prognosis (7 to 31 months). Freedman and Folk7 found that patients with breast cancer had a median survival time <strong>of</strong> 314 days.<br />

They analyzed the patient population with regard to the stage <strong>of</strong> breast cancer.<br />

Patients with Stage 1 or 2 breast cancer with choroidal metastases had a median<br />

survival time <strong>of</strong> 873 days (29.1 months), and patients with Stage 3 or 4 breast<br />

cancer with Choroidal metastases had a median survival time <strong>of</strong> 139 days (4.6<br />

months) 7 . The overall median survival is better in stages I/II than stages III/IV17 .<br />

Conclusions<br />

Due to the earlier recognition <strong>of</strong> cancer and the progress <strong>of</strong> chemotherapeutic<br />

medications, the number <strong>of</strong> patients who manifest Choroidal metastases will<br />

continue to increase. It is essential for the practitioner to be able to recognize this<br />

disease process to institute proper treatment for metastatic disease and prevent<br />

visual loss. A thorough fundus examination, coupled with the use <strong>of</strong> A-scan and<br />

B-scan ultrasonography, will aid in the diagnosis <strong>of</strong> suspicious choroidal lesions.<br />

The goal <strong>of</strong> therapy is <strong>of</strong>ten palliative if the patient is already being treated<br />

systemically for cancer.<br />

References<br />

1. American Cancer Society. Cancer facts and figures 2003. Atlanta: American<br />

Cancer Society, 2003.<br />

2. Ferry AP, Font RL. Carcinoma metastatic to the eye and orbit: a<br />

clinicopathologic study <strong>of</strong> 227 cases. Arch Ophthalmol 1974; 92:276-86.<br />

3. Sheth D, C.A Wasen, D. Schroder, J.E. Boccaccio and L.R .Lloyd, 1999. The<br />

advanced breast biopsy instrumentation (ABBI) experience at a community<br />

hospital. Am. Surg., 65: 726-30.<br />

4. Wiegel T, K.M. Kreusel and N. Bon Feld et al, 1998. Frequency <strong>of</strong><br />

8 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


asymptomatic Choroidal metastases in patient with disseminated breast<br />

cancer: results <strong>of</strong> a prospective screening programme. Br. J. Ophthalmol.,<br />

22: 1159-61.<br />

5. Ratanatharathorn V, Powers WE, Grimm J, et al. eye metastases from<br />

carcinoma <strong>of</strong> breast: diagnosis, radiation treatment and results. Cancer treat<br />

rev 1991; 18:161-76.<br />

6. Shields CL. Plaque radiotherapy for management <strong>of</strong> uveal metastases. Curr<br />

Opin Ophthalmol 1998; 9:31-7.<br />

7. Freedman MI, Folk JC. Metastatic tumors <strong>of</strong> eye and orbit: patient survival<br />

and clinical characteristics. Arch Ophthalmol 1987; 105:1215-9.<br />

8. Demirci H, Shields CL, Chao A, et al. uveal metastases from breast cancer<br />

in 264 patients. Am J Ophthalmol 2003; 136:264-71.<br />

9. Shields CL, Shields JA, Gross N, et al. survey <strong>of</strong> 520 eyes with uveal<br />

metastases. Ophthalmology 1997; 104:1265-76.<br />

10. Brinkley JR Jr. Response <strong>of</strong> Choroidal metastases to multiple drug<br />

chemotherapy. Cancer 1980; 45:1538-39<br />

11. Wilson MW, Czechonska G, Finger PT, et al. chemotherapy for eye cancer.<br />

Surv Ophthalmol 2001; 45:416-44.<br />

12. Rosset A, Zografos L, Coucke P, et al. radiotherapy <strong>of</strong> Choroidal metastases.<br />

Radiotherapy Oncol 1998; 46:263-8.<br />

13. Puri P, Gupta M, Rundle PA, et al. Indocyanine green augmented<br />

transpupillary thermotherapy in the management <strong>of</strong> Choroidal metastases<br />

from breast cancer. Eye 2001; 15(Pt 4): 515-8.<br />

14. Harbour JW. Photodynamic therapy for Choroidal metastases from carcinoid<br />

tumor. Am J Ophthalmol 2004; 137:1143-45.<br />

15. Thatcher N, Thomas PR: Choroidal metastases from breast carcinoma:<br />

a survey <strong>of</strong> 42 patients and the use <strong>of</strong> radiation therapy. Clin Radiol 26:<br />

549–553, 1975.<br />

16. Letson AD, Davidorf FH, Bruce Jr RA: Chemotherapy for treatment <strong>of</strong><br />

Choroidal metastases from breast carcinoma. Am J Opthalmol 93: 102–106,<br />

1982.<br />

17. Freedman MI, Folk JC: Metastatic tumors to the eye and orbit. Patient<br />

survival and clinical characteristics. Arch Opthalmol 105: 1215–1219, 1987<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 9


original article <<br />

Figures<br />

Fig. 1: C/EBPα subgroups and FAB subtypes in our study group.<br />

a b<br />

Fig. 2: Disease free survival (a) and overall survival (b) <strong>of</strong> AML patients based on C/EBPα expression levels.<br />

10 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


notes <<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 11


original article <<br />

Multidrug Resistance- related Protein (MRP) and Lung Resistance Protein (LRP) m<br />

RNA Expression in Egyptian Patients with Acute Leukemia<br />

Amira Ahmed Hammam, MD 1 , Mohsen Mokhtar, MD 2<br />

(1) Department <strong>of</strong> Clinical Pathology, Faculty <strong>of</strong> medicine, Beni Suef University<br />

(2) Department <strong>of</strong> Clinical <strong>Oncology</strong> , Faculty <strong>of</strong> medicine, Cairo University<br />

Corresponding Author: Amira Ahmed Hammam, MD<br />

Lecturer <strong>of</strong> clinical Pathology, Faculty <strong>of</strong> medicine Beni Suef university, Cairo-Egypt<br />

E-mail: amirahammam@gmail.com<br />

Key words: MDR, Multidrug Resistance-related Protein, Lung resistance protein, Prognosis, Acute leukemia.<br />

ISSN: 2070-254X<br />

Abstract<br />

Background and objectives: Clinical resistance to chemotherapy is a major<br />

obstacle in treatment and an important cause <strong>of</strong> death in Acute Leukemia.Such<br />

resistance is usually associated with the expression <strong>of</strong> multidrug resistance(MDR)<br />

genes. The significance <strong>of</strong> MDR genes expression is still controversial<br />

Aim <strong>of</strong> the work: We investigated whether multidrug resistance-related protein<br />

(MRP) and Lung resistance protein (LRP) m RNA expression are associated<br />

with outcomes ,clinical and laboratory findings in acute leukemia patients.<br />

Patients&methods: At diagnosis we examined MRP and LRP m RNA<br />

expression in peripheral blood samples from 50 Egyptian Acute Leukemia<br />

patients (25 myeloid& 25 lymphoblastic) using nested RT-PCR. Ten age matched<br />

normal individuals were included as control group.<br />

Results: m RNA <strong>of</strong> MRP and LRP genes were detected in 28/50 (56%) &<br />

22/50 (44%) respectively, while there was double expression <strong>of</strong> both genes in<br />

18/50 (36%) <strong>of</strong> acute leukemia patients. There was no statistically significant<br />

difference between MRP&LRP positive and negative patients as regards<br />

their clinical and laboratory data including age, sex , hemoglobin, platelets,<br />

hepatosplenomegally, lymphandenopathy and CNS involvement. As regards<br />

Total leucocytic count (TLC) the comparison between positive and negative<br />

cases showed high statistically significant difference (p


National Cancer Institute, Egypt. The median age <strong>of</strong> the patients was 28 years,<br />

with mean 25.3 years (range 2yr-75yr). Diagnosis and classification <strong>of</strong> acute<br />

leukemia was made according to the French-American-British (FAB) criteria<br />

and immunophenotype analyses. There were 25 acute myeloid leukemia (AML)<br />

and 25 acute lymphoblastic leukemia(ALL) patients. Among the 25 AML<br />

patients, two patients had AML M0, 7 M1, 8 M2, 2 M3, 3 M4, 2 M5, 1 M6.<br />

Among the 25 ALL patients, fifteen patients were B-lineage, 6 T-lineage and 4<br />

common ALL.<br />

Ten normal age matched individuals were included as control group<br />

Complete remission(CR)<br />

Complete remission was defined as normocellular bone marrow with less than<br />

5% blasts after induction chemotherapy, no Auer rods, and no evidence <strong>of</strong><br />

extramedullary involvement(Marry et al;2007). Patients who relapsed or died<br />

within 28 days after CR were considered as not having achieved a CR.<br />

Sample collection and Nested RT-PCR assay<br />

Fifty peripheral blood samples collected in EDTA were obtained at the initial<br />

diagnosis. A COR-L23/R cell line was used as a positive control for LRP m<br />

RNA expression, and the HL60/Adr cell line for MRP m RNA expression.<br />

RNAase-free water was used as negative control, also ten age matched normal<br />

individuals were included as control group. Β actin m RNA amplification was<br />

used as an internal control.<br />

Mononuclear cells were separated from peripheral blood samples by density<br />

gradient centrifugation using Ficoll-Hypaque and preserved at -20ºC till use.<br />

Total cellular RNA was extracted from MNCs using a QIAamp RNA blood<br />

kit (Qiagen,Germany) . Complementary DNA (cDNA) was synthesized using<br />

Revert Aid First strand c DNA synthesis kit(Fermentas,K1621). The reaction<br />

was performed at 70ºC for 2 min and 42ºC for 60 min. MRP and LRP m RNA<br />

amplifications were performed after heating the reaction mixture to 99ºC for 5<br />

min.<br />

The first round <strong>of</strong> PCR reactions involved 30 cycles <strong>of</strong>: 1 min denaturing at<br />

94ºC, 1 min annealing at 64ºC and 2 min extension at 72ºC using a thermocycler.<br />

The PCR reactions were carried out in a final volume <strong>of</strong> 25µl containing 12.5 µl<br />

Dream Tag Green PCR Master Mix(Fermentas,K0171 which contains TaqDNA<br />

polymerase in reaction buffer MgCl2 and Dntps), 5/µl <strong>of</strong> 5pmol <strong>of</strong> each primer,<br />

2 µl c DNA, completed to the final volume with nuclease free water. The first<br />

round <strong>of</strong> PCR was followed by a second round <strong>of</strong> 20 cycles. Amplification <strong>of</strong><br />

β-actin m RNA (20 cycle PCR) was performed and the data obtained used to<br />

normalize any variation in samples. PCR primer sequences are shown in Table<br />

1. PCR products were electrophoretically separated on 2% agarose gel and<br />

visualized using ethidium bromide staining. The sample was considered positive<br />

when a clear sharp band was observed at the specific molecular weight;420 bp<br />

for MRP, 239 bp for LRP and 166 bp for β actin. The gel was photographed with<br />

Polaroid film.<br />

Statistics<br />

Data were summarized and presented in the form <strong>of</strong> percentage, range , mean<br />

and median. Descriptive statistics and statistical comparisons were performed<br />

using the statistical s<strong>of</strong>t ware program SPSS (version 14). A P value <strong>of</strong> < 0.05<br />

indicated a significant difference while a p value <strong>of</strong> > 0.05 indicated insignificant<br />

difference.<br />

Table 1: NestedRT-PCR primer sequences for MRP, LRP and β- actin m<br />

RNA amplification (1)<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 13<br />

Results<br />

Expression rates <strong>of</strong> m RNA <strong>of</strong> MRP and LRP genes:<br />

By Nested RT-PCR, m RNA <strong>of</strong> MRP and LRP genes were detected in 28/50<br />

(56%) & 22/50 (44%) respectively, while there was double expression <strong>of</strong> both<br />

genes in 18/50 (36%) <strong>of</strong> acute leukemia patients.<br />

In terms <strong>of</strong> leukemia categories, MRP was expressed in 14/25 (56%) <strong>of</strong> AML<br />

patients and 14/25 (56%) <strong>of</strong> ALL patients; while LRP was expressed in 12/25<br />

(48%) <strong>of</strong> AML Patients and 10/25 (40%) <strong>of</strong> ALL patients.<br />

All normal controls didn’t express both genes.<br />

Comparison between MRP&LRP positive and negative patients regarding their<br />

clinical and laboratory data is presented in table (2).<br />

There was no statistically significant difference between MRP&LRP positive<br />

and negative patients as regards their clinical and laboratory data including age,<br />

sex , hemoglobin, platelets, hepatosplenomegally, lymphandenopathy and CNS<br />

involvement.<br />

As regards Total leucocytic count (TLC) the comparison between positive and<br />

negative cases showed high statistically significant difference (p


original article <<br />

Table 2: Relationship between MRP&LRP genes expression and clinical<br />

and laboratory data <strong>of</strong> patients<br />

Acute leukemia<br />

(n=50)<br />

AML<br />

(n=25)<br />

ALL<br />

(n=25)<br />

Sex<br />

Male(n=24)<br />

Female(n=26)<br />

TLCx10³/cm³<br />

(mean&range)<br />

Hemoglobin gm/dl<br />

(mean&range)<br />

Plateletsx10³/cm³<br />

(mean&range)<br />

MRP<br />

positive negative<br />

28/50 22/50<br />

(56%) (44%)<br />

14/25 11/25<br />

(56%) (44%)<br />

14/25 11/25<br />

(56%) (44%)<br />

17/24 7/24<br />

(70.8%) (29.2%)<br />

12/26 14/26<br />

(46.2%) (53.8%)<br />

90.35* 40.3*<br />

(3-325) (1.35-115)<br />

8.1 7.6<br />

(4.4-11.6) (2.5-10)<br />

98.4 110.6<br />

(8-413) (6-765)<br />

Hepatosplenomegaly 10/28 8/22<br />

(35.7%) (36.4%)<br />

Lymphadenopathy 8/28 7/22<br />

(28.5%) (31.8%)<br />

CNS involvement 4/28 3/22<br />

(14.2%) (13.6%)<br />

Complete Remission 6/28* 15/22*<br />

(21.4%) (68.2%)<br />

*p value <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


RT-PCR the m RNA <strong>of</strong> two genes named MRP&LRP genes. Fifty patients (30<br />

males and 20 females) diagnosed with denovo acute leukemia between January<br />

2009 and December 2009 recruited from the National Cancer Institute, Egypt<br />

were included in this study. The median age <strong>of</strong> the patients was 28 years, with<br />

mean 25.3 years (range 2yr-75yr). Diagnosis and classification <strong>of</strong> acute leukemia<br />

was made according to the French-American-British (FAB) criteria and<br />

immunophenotype analyses. There were 25 acute myeloid leukemia (AML) and<br />

25 acute lymphoblastic leukemia(ALL) patients. Among the 25 AML patients,<br />

two patients had AML M0, 7 M1, 8 M2, 2 M3, 3 M4, 2 M5, 1 M6.<br />

Among the 25 ALL patients, fifteen patients were B-lineage, 6 T-lineage and 4<br />

common ALL.<br />

Ten normal age matched individuals were included in our study as control group.<br />

m RNA <strong>of</strong> MRP and LRP genes were detected in 28/50 (56%) & 22/50 (44%)<br />

respectively, while there was double expression <strong>of</strong> both genes in 18/50 (36%) <strong>of</strong><br />

acute leukemia patients.<br />

In terms <strong>of</strong> leukemia categories, MRP was expressed in 14/25 (56%) <strong>of</strong> AML<br />

patients and 14/25 (56%) <strong>of</strong> ALL patients; while LRP was expressed in 12/25<br />

(48%) <strong>of</strong> AML Patients and 10/25 (40%) <strong>of</strong> ALL patients.<br />

All normal controls didn’t express both genes.<br />

There was no statistically significant difference between MRP&LRP positive<br />

and negative patients as regards their clinical and laboratory data including age,<br />

sex , hemoglobin, platelets, hepatosplenomegally, lymphandenopathy and CNS<br />

involvement.<br />

Mary M. et al (2007)(2) showed that the m RNA expression <strong>of</strong> resistance genes<br />

were not significantly associated with the age <strong>of</strong> patients.<br />

As regards Total leucocytic count (TLC) the comparison between positive and<br />

negative cases showed high statistically significant difference (p


original article <<br />

resistance <strong>of</strong> tumor cells. Anticancer Res; 20:4261-74<br />

9. Kakihara T, Tanaka A, Watanabe A, Yamamoto K, Kanto K, Kataoka S,<br />

Ogawa A, Asami K, Uchiyama M (1999): Expression <strong>of</strong> multidrug resistance-related<br />

genes does not contribute to risk factors in newly diagnosed<br />

childhood acute lymphoblastic leukemia . Pediatr Int ; 41:641-7<br />

10. Dhooge C, De Moerloose B, Laureys G, Kint J et al (1999): P-glycoprotein<br />

is an independent prognostic factor predicting relapse in childhood acutelymphoblastic<br />

leukemia: results <strong>of</strong> a 6-year prospective study. Br J Haematol;<br />

105:676-83<br />

11. Valera E.T, Scrideli C.A, Queiroz R.G, Mari B.M.O,Tone L.G (2004): Multiple<br />

drug resistance protein (MDR-1), multidrug resistance related protein<br />

(MRP) and lung resistance protein (LRP) gene expression in childhood<br />

acute lymphoblastic leukemia. Sao Paulo Med J ;122(4):166-71<br />

12. Klein I., Sarkadi B. and Varadi A (1999): An inventory <strong>of</strong> the human ABC<br />

proteins. Biochim.Biophys.Acta; 1461:237-262<br />

13. Borst P., Evers R., Kool M., and Wijniholds J. (2000): A family <strong>of</strong> drug<br />

transporters: the multidrug-resistance-associated proteins.J.Natl.Cancer<br />

Inst.(Bethesda),92:1295-1302<br />

14. Burger H.,Foekens J.A.,Look M.P, Gelder M., Klijn J. G.M., Wiemer<br />

E.A.C, Stoter G.,and Nooter K. (2003): RNA Expression <strong>of</strong> Breast Cancer<br />

R esistance Protein,Lung Resistance-related Protein, Multidrug Resistanceassociated<br />

Proteins 1 and 2, and Multidrug Resistance Gene 1 in Breast<br />

Cancer: Correlation with chemotherapeutic Response. Clinical Cancer Research<br />

vol. 9:827-836<br />

15. Hipfner D.R., Deeley R.G., and Cole S.P.C (1999): Structural mechanistic<br />

and clinical aspects <strong>of</strong> MRP1. Biochim.Biophys.Acta,1461:359-376<br />

16. Larsen A.K., Escargueil A.E., and Skladanowski A. (2000): Resistance<br />

mechanisms associated with altered intracellular distribution <strong>of</strong> anticancer<br />

agents.Phamacol.Ther;85:217-229<br />

17. Scheffer G.L., Schroeijers A.B., Izquierdo M.A., Wiemer E.A., and Scheper<br />

R.J.(20000): Lung resistance-related protein/majir vault protein and vaults<br />

in multidrug-resistance cancer.Curr.opin.oncol;12:550-556<br />

18. Sauerbrey A, Voigt A, Wittig S, Hafer R, Zintl F (2002): Messenger RNA<br />

analysis <strong>of</strong> the multidrug resistance related protein (MRP1) and the lung<br />

resistance protein (LRP) in de novo and relapsed childhood acute lymphoblastic<br />

leukemia. Leuk Lymphoma; 43(4):875-9<br />

19. Plasschaert S.L, Vellenga E, de Bont E.S, et al (2003): High functional pglycoprotein<br />

activity is more <strong>of</strong>ten present in T-cell acute lymphoblastic leukemia<br />

cells in adults than in children. Leuk Lymphoma; 44(1):85-95<br />

20. den Boer M.L, Pieters R, Karzemier K.M, et al (1998): The modulation<br />

effect <strong>of</strong> PSC 833,Cyclosporin A, Verapamil and genistein on in vitro cytotoxicity<br />

and intracellular content <strong>of</strong> daunorubicin in childhood acute lymphoblastic<br />

leukemia. Leukemia; 12(6):912-20<br />

16 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


notes <<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 17


original article <<br />

Phase II pilot study <strong>of</strong> weekly Docetaxel as Neoadjuvant Chemotherapy for operable<br />

Breast Cancer<br />

El. Bassiouny M. Mohamed, MD 1 , Hesham El. Ghazaly, MD 1 , Mahmoud A. El. Lithy, MD 1 , Ramy R. Ghali, MD 1 , Moustafa<br />

Mahmoud, MD 2 , Gamal Fawzy MD and Osama Mahmoud MD, FRCS 3<br />

(1) Department <strong>of</strong> Radiation <strong>Oncology</strong> and Nuclear Medicine, Ain Shams University.<br />

(2) Department <strong>of</strong> Radiology, Ain Shams University.<br />

(3) Department <strong>of</strong> General Surgery, Ain Shams University.<br />

Corresponding Author: Mahmoud Ellithy, MD<br />

Address: Ebadelrahman city, Ring road. Omar Ebn Elkkattab st. No 58.<br />

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

Key words: Weekly, Docetaxel, Neoadjuvant, Breast cancer.<br />

ISSN: 2070-254X<br />

Abstract<br />

Purpose: In this study <strong>of</strong> weekly docetaxel in the neoadjuvant treatment <strong>of</strong> stage<br />

II breast cancer, we evaluated the efficacy and safety <strong>of</strong> docetaxel and analyzed<br />

the correlation between the response observed and the expression <strong>of</strong> c-erbB2,<br />

ER status.<br />

Patients and Methods: This study included patients with previously untreated,<br />

stage II & III breast cancer. Docetaxel was given as a 30-min i.v. infusion at<br />

a dose <strong>of</strong> 40 mg/m2 weekly for 6 weeks, followed by 2 weeks rest. Patients<br />

received two 8-week cycles <strong>of</strong> treatment. Patients achieving a CR or partial<br />

response or who had stable disease at the end <strong>of</strong> treatment proceeded directly<br />

to surgery.<br />

Results: A total <strong>of</strong> 40 patients were evaluated by intention-to-treat analysis<br />

for efficacy and safety. The overall clinical response rate was 65% (complete<br />

and partial response, 25 and 40%, respectively). Six patients (15%) achieved<br />

a pathological complete response. There was no correlation between response<br />

to docetaxel and the expression <strong>of</strong> molecular markers, however, the majority<br />

<strong>of</strong> the pathological complete responses were observed in patients with<br />

c-erbB2-negative tumors. Nonhematological toxicity was more common than<br />

hematological toxicity, with alopecia and asthenia were the most frequently<br />

reported adverse events. Severe hematological toxicity was rare.<br />

Conclusion: Weekly docetaxel appears to be very effective in the neoadjuvant<br />

setting. A high pathological response rate was achieved with tolerable toxicity.<br />

Introduction<br />

Breast cancer is the most common cancer in women and it is incurable when<br />

metastases are diagnosed. Taxanes, namely docetaxel and paclitaxel, are effective<br />

chemotherapeutic agents in the metastatic, neoadjuvant and adjuvant settings.<br />

HER-2 over expression is detected in 25–30% <strong>of</strong> breast cancer , it confers<br />

aggressive tumor behavior as well as resistance to some systemic treatments and<br />

has been associated with poor outcome, in both node-negative and node-positive<br />

early breast cancer [1-4]. In vitro, HER-2 overexpression confers increased<br />

resistance to paclitaxel in breast cancer cells, while HER-2 degradation increases<br />

docetaxel-induced apoptosis [5-7]. This is further supported by data from a phase<br />

III clinical trial showing that paclitaxel response rate was significantly improved<br />

in breast cancer patients when HER-2 was downregulated by the humanized anti-<br />

HER-2 antibody, trastuzumab [8].<br />

Neoadjuvant therapy for breast cancer generally refers to the administration<br />

<strong>of</strong> chemotherapy prior to local treatment with surgery and/or radiation. The<br />

biological rationale for neoadjuvant therapy <strong>of</strong> breast cancer is based on the<br />

observation <strong>of</strong> accelerated metastatic growth following tumor resection..<br />

Neoadjuvant chemotherapy minimize the emergence <strong>of</strong> chemo resistant clones<br />

,increasing tumor resectability by reducing the size <strong>of</strong> an unresectable tumor,<br />

improving local control and improving cosmesis by allowing breast-conserving<br />

treatment. It also <strong>of</strong>fers an important test bed for novel therapies including new<br />

drugs and new combinations <strong>of</strong> drugs. More recently, neoadjuvant therapy has<br />

been studied as a way <strong>of</strong> testing the relevance <strong>of</strong> biological markers in predicting<br />

disease outcome. The expression <strong>of</strong> the proto-oncogenes c-erbB2 (her2/neu),<br />

bcl-2 and p53 have been evaluated as predictive markers in several trials without<br />

clearly defined results [9-12].<br />

Observations from early studies, both single arm phase II studies and those<br />

randomizing pre-operative systemic therapy against post-operative adjuvant<br />

therapy, have confirmed that those women whose tumors have a pathological<br />

complete response to neo-adjuvant chemotherapy have the best long-term<br />

outcome, and this remains true after multivariate analysis [13]. It has even been<br />

suggested that regimens achieving a higher proportion <strong>of</strong> patients in pathological<br />

complete response should then be used in the adjuvant setting as they must give<br />

a survival improvement on older treatments [14].<br />

The taxanes have emerged as critically important drugs in the treatment <strong>of</strong><br />

patients with breast cancer. Taxanes were generally recognized as evidencebased<br />

components <strong>of</strong> therapy for metastatic breast cancer within a few short<br />

years <strong>of</strong> their initial phase II evaluations. In addition, the data in support <strong>of</strong> their<br />

use in the adjuvant and neoadjuvant settings continue to strengthen [15].<br />

Docetaxel is a taxane prepared by semisysnthesis beginning with a precursor<br />

extracted from the needles <strong>of</strong> yew plant it binds to the ß-tubulin subunit causing<br />

inhibition <strong>of</strong> microtubule depolemerization [16], and several phase II and III<br />

trials reported a high activity in first- and second-line therapy <strong>of</strong> metastatic breast<br />

cancer, as well as in patients previously exposed or resistant to anthracycline [17-<br />

18 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


18]. When docetaxel was compared with doxorubicin, it produced statistically<br />

superior response rates compared with doxorubicin (48% versus 33%, P = 0.008)<br />

and longer time to treatment failure [17]. It has also been studied in patients who<br />

have progressed on anthracycline-containing chemotherapy and differences in<br />

progression-free survival have been found [18].<br />

Weekly regimens <strong>of</strong> taxanes may enhance dose intensity by minimizing regrowth<br />

<strong>of</strong> cells between cycles <strong>of</strong> treatment and can also be combined with other new<br />

drugs such as biological agents directed against the tyrosine kinase I receptor<br />

c-erbB2 [19] . Experience with docetaxel as neoadjuvant chemotherapy has<br />

mainly been in combination with an anthracycline . The rates <strong>of</strong> pahological<br />

complete remission (pCR) reported in these trials are similar to those obtained<br />

with standard regimens, although the best regimen with docetaxel as neoadjuvant<br />

chemotherapy has not yet been defined.<br />

Based on indirect comparisons as well as the results <strong>of</strong> the recent randomized<br />

trial conducted in patients with metastatic breast cancer, docetaxel appears to<br />

be the more active taxane. In addition to its longer half-life, docetaxel also has a<br />

more rapid cellular uptake and longer intracellular retention than paclitaxel [20]..<br />

Docetaxel is highly active when given as a short, intermittent infusion. [21].<br />

This study is a pilot phase II trial <strong>of</strong> single-agent docetaxel given on a weekly<br />

schedule as neoadjuvant treatment for patients with breast cancer. Our primary<br />

aim was to measure the overall objective clinical response rate to weekly<br />

docetaxel. Secondary objectives included assessing the safety <strong>of</strong> the regimen,<br />

measuring the pathological response rate to docetaxel, and evaluating the role<br />

<strong>of</strong> the c-erbB2 receptor and ER as potential predictive factors <strong>of</strong> response to<br />

single-agent docetaxel.<br />

Patients and Methods<br />

Patient Population<br />

Patients with previously untreated, histologically confirmed, operable breast<br />

cancer were included in the study. Infiltrating disease was confirmed before<br />

chemotherapy for all patients. Patients were required to have measurable<br />

disease by physical examination and diagnostic breast imaging (mammogram,<br />

ultrasound and Magnetic Resonant Image) with a primary tumor ≥ 2 cm. All<br />

patients were ≥ 18 years. Patients had to have Eastern Cooperative <strong>Oncology</strong><br />

Group performance status < 2, hemoglobin ≥ 10 g/dl, neutrophils ≥ 2 × 109 /<br />

liter, and platelets ≥ 100 × 109 /liter, with adequate hepatic and renal function<br />

(including calculated creatinine clearance ≥ 60 ml/min). Cardiac function was<br />

evaluated by echocardiography and a left ventricular ejection fraction <strong>of</strong> ≥50%<br />

was required. Patients were excluded if they had bilateral tumor or metastatic<br />

disease as confirmed by chest radiography, liver ultrasound or computer<br />

tomography imaging and bone scintigraphy, or if they had any other severe or<br />

uncontrolled systemic disease. Fertile women had to have a negative pregnancy<br />

test and had to be using adequate, non hormonal contraception.<br />

Treatment<br />

Docetaxel was given as a 30-min i.v. infusion at a dose <strong>of</strong> 40 mg/m2 weekly<br />

for 6 weeks, followed by 2 weeks rest. Patients received two 8-week cycles <strong>of</strong><br />

treatment. Patients achieving a CR or partial response or who had stable disease<br />

at the end <strong>of</strong> treatment proceeded directly to surgery. After surgery, adjuvant<br />

chemotherapy was delivered according to the standard regimen.<br />

Docetaxel administration was delayed for up to 1 week in the event <strong>of</strong> a neutrophil<br />

count < 1 × 109 /liter, platelet count 10% tumor cells; and score 3+,<br />

strong, complete membrane staining in >10% <strong>of</strong> tumor cells. Scores 0 and 1+<br />

were considered negative, and scores 2+ and 3+ were considered positive for<br />

c-erbB2 overexpression.<br />

Statistical Analysis<br />

The sample size was calculated using with a type I error <strong>of</strong> 5% and a study power<br />

<strong>of</strong> 80%. The target enrollment was estimated to be 40 patients. All patients who<br />

fulfilled the inclusion criteria and received at least one infusion were evaluated<br />

for efficacy and safety on an intention-to-treat analysis. The statistical analysis<br />

was performed by SPSS version 17. All variables were analyzed by descriptive<br />

methods. Chi square test was used to assess the relation between response to<br />

Docetaxel (clinical and pathological response) with Her 2 neu expression and<br />

Hormonal receptors status.<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 19


original article <<br />

Results<br />

Patients<br />

• Forty patients were included in the trial. Characteristics <strong>of</strong> the<br />

patients are summarized in Table 1. All patients were evaluated for<br />

efficacy and safety on an intention-to-treat basis. Two patients were<br />

withdrawn with progressive disease during treatment.<br />

Efficacy<br />

Response rates is illustrated in table 2 .Before treatment, only 20 (50%) patients<br />

were eligible for lumpectomy, and no patient initially scheduled for lumpectomy<br />

subsequently required mastectomy. The precise surgical procedure carried out<br />

was decided upon by the surgeon on a case-by-case basis, with consent from the<br />

patient. A pCR, with no evidence <strong>of</strong> invasive tumor in breast and lymph nodes,<br />

was confirmed in 10 <strong>of</strong> 40 patients (25%), including 2 patients with residual<br />

carcinoma in situ only.<br />

Immunohistochemistry<br />

Tumor paraffin blocks were obtained from 40 patients before treatment.<br />

No correlation was seen between clinical or pathological response with the<br />

expression <strong>of</strong> any <strong>of</strong> the biological markers assessed, however, the majority <strong>of</strong><br />

the pCRs were observed in patients with c-erbB2-negative tumors. (Table 3)<br />

Safety<br />

Toxicity was assessed in 40 patients (Table 4).<br />

Discussion<br />

In this study the overall response rate was slightly low (65%) compared with<br />

other recent studies , the pCR rate was very high with 10 patients (25%) showing<br />

no evidence <strong>of</strong> invasive tumor at definitive surgery.<br />

Most <strong>of</strong> the neoadjuvant trials reported to date with docetaxel are Phase II<br />

studies in combination with an anthracycline and no particular combination or<br />

regimen appears to be outstanding in terms <strong>of</strong> pathological and clinical response<br />

rates. Trial B-27 from the National Surgical Adjuvant Breast and Bowel Project<br />

(NSABP) demonstrated that the addition <strong>of</strong> four cycles <strong>of</strong> sequential, preoperative<br />

docetaxel to preoperative AC(Adriamycin /cyclophosphamide) chemotherapy in<br />

2411 patients produced a significantly superior outcome ( Overall response rate<br />

(ORR)=90.7% ,pCR=26.3%). relative to four cycles <strong>of</strong> AC alone (ORR=85.7%<br />

,pCR=12.9%). [24].<br />

Some have criticized the design <strong>of</strong> the NSABP B-27 trial because the<br />

preoperative regimens were <strong>of</strong> different durations (four versus eight cycles)<br />

and suggested that the favorable results in the AC/docetaxel arm may be due to<br />

the delivery <strong>of</strong> additional cycles <strong>of</strong> chemotherapy rather than a distinct taxane<br />

benefit. However, the results <strong>of</strong> a study conducted at the University <strong>of</strong> Aberdeen,<br />

in which eight cycles <strong>of</strong> neoadjuvant CVAP (cyclophosphamide/vincristine/<br />

doxorubicin/prednisolone) chemotherapy were compared with four cycles <strong>of</strong><br />

CVAP followed by four cycles <strong>of</strong> docetaxel (eight cycles total) prior to surgery<br />

, suggested that the addition <strong>of</strong> the taxane is indeed beneficial (ORR=66%<br />

vs 94% ,pCR= 16% vs 34% respectively). [25]. In that trial, 162 patients with<br />

large or locally advanced breast cancer were randomized to the study regimens,<br />

and 145 patients completed eight cycles <strong>of</strong> neoadjuvant therapy.. The results<br />

were statistically significant in the primary analysis and in the intent-to-treat<br />

population. Furthermore, two patients who received eight cycles <strong>of</strong> CVAP<br />

developed progressive disease after initially responding to the first four cycles<br />

<strong>of</strong> CVAP, which suggests the development <strong>of</strong> acquired resistance to the regimen.<br />

This observation was not seen in the docetaxel group, and this finding supports<br />

the use <strong>of</strong> non-cross-resistant chemotherapy combinations such as anthracyclines<br />

plus taxanes.<br />

The NSABP B-27 data are corroborated by the results <strong>of</strong> the GEPARDUO trial<br />

conducted by the German Adjuvant Breast Cancer Group, which compared four<br />

cycles <strong>of</strong> dose-dense AT (doxorubicin/docetaxel) given concomitantly with a<br />

sequential regimen <strong>of</strong> four cycles <strong>of</strong> doxorubicin/cyclophosphamide followed<br />

by four cycles <strong>of</strong> docetaxel (AC-T) as neoadjuvant therapy in 913 patients with<br />

operable breast carcinoma [26]. In the primary analysis, AC-T was associated<br />

with a superior pCR rate and ORR, as well as a greater rate <strong>of</strong> breast-conserving<br />

surgery and higher incidence <strong>of</strong> pathologically negative axillary lymph nodes (<br />

ORR=77.2% vs 86.8% ,pCR= 11.5% vs.22.4% respectively).<br />

Conversely, interim results <strong>of</strong> a phase III trial conducted by the Anglo-Celtic<br />

Cooperative <strong>Oncology</strong> Group showed that there was no benefit to using a<br />

concomitant docetaxel/doxorubicin regimen every 3 weeks, relative to AC,<br />

in the neoadjuvant setting [27]. A total <strong>of</strong> 363 women with locally advanced<br />

breast cancer were randomized in that trial. At the time <strong>of</strong> analysis, there was a<br />

trend toward a higher ORR with AT (72% versus 62%; p = 0.07), but there were<br />

no differences in rates <strong>of</strong> pCR, axillary lymph node involvement, relapse-free<br />

survival, or overall survival.<br />

In our study, using single-agent docetaxel in a weekly schedule, resulted in a<br />

high number <strong>of</strong> pathological responses and a favorable toxicity pr<strong>of</strong>ile. The fact<br />

that the proportion <strong>of</strong> stage II patients included was high (85%), with a relatively<br />

small median tumor size (4.6 cm), may have favorably affected this pCR.<br />

Docetaxel was generally well tolerated. Apart from alopecia, asthenia was the<br />

most frequent side effect but did not result in patient withdrawal from the study.<br />

Nail disorders and acral erythema were frequent, although the number <strong>of</strong> patients<br />

with grade 3–4 events was low, and the symptoms were reversible. Grade 3–4<br />

myelosuppression was rare in our study, with only 2 cases <strong>of</strong> neutropenia and<br />

none <strong>of</strong> thrombocytopenia.<br />

In neoadjuvant trials, HER-2 positive was associated with an improved response<br />

to dose-dense paclitaxel in 15 <strong>of</strong> 21 stage T2–3 BC patients; clinical response<br />

was more than double in patients with HER2-positive tumors treated with<br />

paclitaxel (P < 0.05). Although this trial was prospective, it was not randomized,<br />

had a small number <strong>of</strong> patients and has been published only as an abstract [28].<br />

Another small prospective study failed to demonstrate any correlation between<br />

HER-2 positivity and pathological complete response (pCR) in 29 patients with<br />

locally advanced breast cancer (LABC), T3 or T4, treated with doxorubicin<br />

followed by paclitaxel or paclitaxel followed by doxorubicin in a dose-dense<br />

regimen [29]. These negative results were also confirmed using FISH (Her-2/<br />

CEP17 >2) in 71 patients treated with neoadjuvant paclitaxel or docetaxel given<br />

every 3 weeks [30]. Estévez et al. [31] also failed to demonstrate correlation<br />

between pathological or clinical response and HER-2 expression (P = 0.355 and<br />

P = 0.942, respectively) in 56 stage II–III breast cancer patients treated with<br />

weekly neoadjuvant docetaxel (ORR 68% and pCR 16%). However, all these<br />

three trials were non-randomized, had few patients and in two studies, HER-2<br />

status was not confirmed by FISH.<br />

Interesting findings from Modi et al. [32] showed that phosphorylated-activated<br />

HER-2 is associated with clinical resistance to taxanes in 126 patients enrolled in<br />

different trials with single-agent taxanes for metastatic and, perhaps, functional<br />

assessment <strong>of</strong> HER-2 status may provide unique predictive information not seen<br />

with conventional assessment.<br />

More recently, gene expression pr<strong>of</strong>iling techniques have been used for the<br />

development <strong>of</strong> a prediction model for response to docetaxel and paclitaxel.<br />

20 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


Chang et al. [33] have reported 92 genes that correlated with docetaxel response<br />

(P = 0.001) using microarray technology. Sensitive tumors had higher expression<br />

<strong>of</strong> genes involved in cell cycle, cytoskeleton, adhesion, protein transport, protein<br />

modification, transcription and stress or apoptosis; whereas resistant tumors<br />

showed increased expression <strong>of</strong> some transcriptional and signal transduction<br />

genes. However, this study was not designed to discover specific genes for<br />

docetaxel response or resistance, but rather to identify patterns <strong>of</strong> many genes<br />

that could be used as a predictive test in patients with breast cancer. [34].Ayers et<br />

al. [35] have examined the feasibility <strong>of</strong> developing a multigene predictor <strong>of</strong> pCR<br />

to sequential weekly paclitaxel and FAC (T/FAC) neoadjuvant CT for breast<br />

cancer. pCR was achieved in 13 patients (31%) out <strong>of</strong> 42 patients: 24 patients<br />

were used in the training set and 18 patients in the validation set. The authors<br />

could not identify any single marker that was sufficiently associated with pCR to<br />

be used as an individual predictor. [36].<br />

In our study, we found no association between overexpression <strong>of</strong> c-erbB2 and<br />

the clinical and pathological response to neoadjuvant docetaxel. However, the<br />

majority <strong>of</strong> the pCRs were observed in patients with c-erbB2-negative tumors.<br />

The number <strong>of</strong> the patients in our study was low, and this trend should be<br />

studied in a larger trial. In addition, these data suggest that blocking c-erbB2<br />

overexpression using a monoclonal antibody such as trastuzumab (Herceptin)<br />

could enhance the response to docetaxel.<br />

This pilot phase II trial highlights the need to identify the best schedule for<br />

neoadjuvant docetaxel. In our study, single-agent docetaxel given on a weekly<br />

schedule appeared to be very effective and well tolerated as neoadjuvant<br />

chemotherapy, with a high documented pCR.<br />

Because at present there is no evidence that primary systemic therapy <strong>of</strong>fers a<br />

disease-free survival or overall survival benefit over adjuvant systemic therapy,<br />

knowledgeable patients may choose to receive systemic therapy before surgical<br />

resection to take advantage <strong>of</strong> the response-assessment <strong>of</strong> the primary tumor<br />

before it is removed.<br />

Acknowledgment<br />

We are in debited to all the nursing staff, the residents and the patients who<br />

accepted to join this study in the Clinical <strong>Oncology</strong> Department, Faculty <strong>of</strong><br />

Medicine, Ain ShamsUniversity.<br />

References<br />

1. Ross JS, Fletcher JA, Linette GP, et al: The Her-2/neu gene and protein in<br />

breast cancer 2003: biomarker and target <strong>of</strong> therapy. Oncologist 8:307–325,<br />

2003<br />

2. Joensuu H, Isola J, Lundin M, et al: Amplification <strong>of</strong> erbB2 and erbB2<br />

expression are superior to estrogen receptor status as risk factors for distant<br />

recurrence in pT1N0M0 breast cancer: a nationwide population-based<br />

study. Clin Cancer Res 9:923–930, 2003<br />

3. Tetu B, Brisson J : Prognostic significance <strong>of</strong> HER-2/neu oncoprotein<br />

expression in node-positive breast cancer. The influence <strong>of</strong> the pattern <strong>of</strong><br />

immunostaining and adjuvant therapy. Cancer 73:2359–2365, 1994<br />

4. Kronqvist P, Kuopio T, Nykanen M, et al: Predicting aggressive outcome in<br />

T1N0M0 breast cancer. Br J Cancer 91:277–281, 2004<br />

5. Yu D, Liu B, Tan M, et al : Overexpression <strong>of</strong> c-erbB-2/neu in breast<br />

cancer cells confers increased resistance to Taxol via mdr-1-independent<br />

mechanisms. Oncogene 13:1359–1365, 1996<br />

6. Witters LM, Santala SM, Engle L, et al: Decreased response to paclitaxel<br />

versus docetaxel in HER-2/neu transfected human breast cancer cells. Am J<br />

Clin Oncol 26:50–54, 2003<br />

7. Bali P, Pranpat M, Swaby R, et al: Activity <strong>of</strong> suberoylanilide hydroxamic<br />

acid against human breast cancer cells with amplification <strong>of</strong> her-2. Clin<br />

Cancer Res 11:6382–6389, 2005<br />

8. Slamon DJ, Leyland-Jones B, Shak S, et al: Use <strong>of</strong> chemotherapy plus<br />

a monoclonal antibody against HER2 for metastatic breast cancer that<br />

overexpresses HER2. N Engl J Med 344:783–792, 2001<br />

9. Archer S. G., Eliopoulos A., Spandidos D., etal: Expression <strong>of</strong> ras p21, p53,<br />

and c-erbB-2 in advanced breast cancer and response to first line hormonal<br />

therapy. Br. J. Cancer, 72: 1259-1266, 1995.<br />

10. Chevillard S., Pouillart P., Beldjord C., etal: Sequential assessment <strong>of</strong><br />

multidrug resistance phenotype and measurements <strong>of</strong> S-phase fraction as<br />

predictive markers <strong>of</strong> breast cancer response to neoadjuvant chemotherapy.<br />

Cancer (Phila.), 77: 292-300, 1996.<br />

11. Makris A., Powles T. J., Dowsett M., etal:Prediction <strong>of</strong> response to<br />

neoadjuvant chemoendocrine therapy in primary breast carcinoma. Clin.<br />

Cancer Res., 3: 593-600, 1997.<br />

12. Chang J., Powles T. J., Allred D. C., etal: Biologic markers as predictors <strong>of</strong><br />

clinical outcome from systemic therapy for primary operable breast cancer.<br />

J. Clin. Oncol., 17: 3058-3063, 1999.<br />

13. Bonadonna G., Valagussa P., Brambilla C., etal: Primary chemotherapy in<br />

operable breast cancer: eight-year experience at the Milan Cancer Institute.<br />

J. Clin. Oncol., 16: 93-100, 1998<br />

14. Fisher B, Bryant J, Wolmark N, et al: Effect <strong>of</strong> preoperative chemotherapy<br />

on the outcome <strong>of</strong> women with operable breast cancer. J Clin Oncol 16:<br />

2672–2685, 1998<br />

15. Miller KD, Sledge GW Jr. Taxanes in the treatment <strong>of</strong> breast cancer: a<br />

prodigy comes <strong>of</strong> age. Cancer Invest 17: 121–136, 1999<br />

16. Ringel I, Horwitz SB : Studies with RP 56976 (Taxotene): a semi-synthetic<br />

analog <strong>of</strong> Taxol. J Natl Cancer Inst 83: 288–291, 1991<br />

17. Chan S, Friedrichs K, Noel D, et al: Prospective randomized trial <strong>of</strong><br />

docetaxel versus doxorubicin in patients with metastatic breast cancer. J<br />

Clin Oncol 17: 2341–2354, 1999<br />

18. Nabholtz JM, Senn HJ, Bezwoda WR, et al: Prospective randomized trial <strong>of</strong><br />

docetaxel versus mitomycin C plus vinblastine in patients with metastatic<br />

breast cancer progressing despite previous anthracycline-containing<br />

chemotherapy. J Clin Oncol 17: 1413–1424, 1999<br />

19. Norton L. Theoretical concepts and the emerging role <strong>of</strong> taxanes in adjuvant<br />

therapy. Oncologist, 6 (Suppl. 3): 30-35, 2001.<br />

20. Riou JF, Petitgenet O, Combeau C, et al: Cellular uptake and efflux <strong>of</strong><br />

docetaxel (Taxotere ® ) and paclitaxel (Taxol ® ) in P388 cell line. Proc Am<br />

Assoc Cancer Res 35:385, 1994<br />

21. Ravdin P, Erban J, Overmoyer B et al. Phase III comparison <strong>of</strong> docetaxel<br />

and paclitaxel in patients with metastatic breast cancer. Eur J Cancer 1(suppl<br />

5):S201, 2003<br />

22. Miller A. B., Hoogstraten B., Staquet M., Winkler A : Reporting results <strong>of</strong><br />

cancer treatment. Cancer (Phila.), 47: 207-214, 1981.<br />

23. National Cancer Institute. . Guidelines for the Reporting <strong>of</strong> Adverse Drugs<br />

Reactions, Division <strong>of</strong> Cancer Treatment Bethesda, MD 1988.<br />

24. Bear HD, Anderson S, Brown A, et al: The effect on tumor response <strong>of</strong><br />

adding sequential preoperative docetaxel to preoperative doxorubicin and<br />

cyclophosphamide: preliminary results from National Surgical Adjuvant<br />

Breast and Bowel Project Protocol B-27. J Clin Oncol 21:4165–4174, 2003<br />

25. Smith IC, Heys SD, Hutcheon AW, et al: Neoadjuvant chemotherapy in<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 21


original article <<br />

breast cancer: significantly enhanced response with docetaxel. J Clin Oncol<br />

20:1456–1466, 2002<br />

26. von Minckwitz G, Raab G, Schuette M, et al: Dose-dense versus sequential<br />

adriamycin/docetaxel combination as preoperative chemotherapy (pCHT)<br />

in operable breast cancer (T2-3, N0-2, M0)—primary endpoint analysis <strong>of</strong><br />

the GEPARDUO-study. Proc Am Soc Clin Oncol 21:43a, 2002<br />

27. Evans TRJ, Gould J, Foster E, et al: Phase III randomized trial <strong>of</strong> adriamycin<br />

(A) and docetaxel (D) versus A and cyclophosphamide (C) as primary<br />

medical therapy (PMT) in women with breast cancer: an ACCOG study.<br />

Proc Am Soc Clin Oncol 21:35a, 2002<br />

28. Volm M, Herma, Symmans WF, et al: Her2 status predicts response to<br />

preoperative paclitaxel in patients with breast cancer. Proc Am Soc Clin<br />

Oncol, 1999 (suppl ; abstr 394).<br />

29. Stearns V, Singh B, Tsangaris T, et al: A prospective randomized pilot study<br />

to evaluate predictors <strong>of</strong> response in serial core biopsies to single agent<br />

neoadjuvant doxorubicin or paclitaxel for patients with locally advanced<br />

breast cancer. Clin Cancer Res 9:124–133, 2003<br />

30. Gonzalez-Angulo AM, Krishnamurthy S, Yamamura Y, et al: Lack <strong>of</strong><br />

association between amplification <strong>of</strong> her-2 and response to preoperative<br />

taxanes in patients with breast carcinoma. Cancer 101:258–263, 2004<br />

31. Estevez LG, Cuevas JM, Anton A, et al: Weekly docetaxel as neoadjuvant<br />

chemotherapy for stage II and III breast cancer: efficacy and correlation<br />

with biological markers in a phase II, multicenter study. Clin Cancer Res<br />

9:686–692, 2003<br />

32. Modi S, DiGiovanna MP, Lu Z, et al: Phosphorylated/activated HER2 as<br />

a marker <strong>of</strong> clinical resistance to single agent taxane chemotherapy for<br />

metastatic breast cancer. Cancer Invest 23:483–487, 2005<br />

33. Chang JC, Wooten EC, Tsimelzon A, et al: Gene expression pr<strong>of</strong>iling for<br />

the prediction <strong>of</strong> therapeutic response to docetaxel in patients with breast<br />

cancer. Lancet 362:362–369, 2003<br />

34. Iwao-Koizumi K, Matoba R, Ueno N, et al: Prediction <strong>of</strong> docetaxel response<br />

in human breast cancer by gene expression pr<strong>of</strong>iling. J Clin Oncol ( 23:422–<br />

431, 2005<br />

35. Ayers M, Symmans WF, Stec J, et al: Gene expression pr<strong>of</strong>iles predict<br />

complete pathologic response to neoadjuvant paclitaxel and fluorouracil,<br />

doxorubicin, and cyclophosphamide chemotherapy in breast cancer. J Clin<br />

Oncol 22:2284–2293, 2004<br />

36. Hess KR, Anderson K, Symmans WF, et al: Pharmacogenomic predictor <strong>of</strong><br />

sensitivity to preoperative chemotherapy with paclitaxel and fluorouracil,<br />

doxorubicin, and cyclophosphamide in breast cancer. J Clin Oncol 24:4236–<br />

4244, 2006<br />

22 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org<br />

Tables<br />

Table 1. Baseline characteristics <strong>of</strong> patients<br />

No. <strong>of</strong> patients (%)<br />

Total no. <strong>of</strong> patients<br />

Age (yr)<br />

40 (100)<br />

Mean<br />

53±SD19.7<br />

Range<br />

Tumor size (cm)<br />

28–73<br />

Mean<br />

4.4±SD1.7<br />

Range<br />

ECOG<br />

2–8.5<br />

a performance status<br />

0<br />

39 (97.5)<br />

1<br />

Menopausal status<br />

1 (2.5)<br />

Premenopausal<br />

18 (45)<br />

Perimenopausal<br />

2 (5)<br />

Postmenopausal<br />

Disease stage<br />

20 (50)<br />

IIA<br />

16 (40)<br />

T2N0M0<br />

16 (40)<br />

IIB<br />

18 (45)<br />

T3N0M0<br />

14 (35)<br />

T2N1M0<br />

4 (10)<br />

IIIA<br />

4 (10)<br />

T2N2M0<br />

2 (5)<br />

T3N1M0<br />

2 (5)<br />

IIIB<br />

2 (5)<br />

T3N2M0<br />

aEastern Cooperative <strong>Oncology</strong> Group<br />

2 (5)<br />

Table 2: Response rate to docetaxel on intention-to-treat basis<br />

No. <strong>of</strong> patients %<br />

CR 10 25<br />

Partial response 16 40<br />

Stable disease 12 30<br />

Progressive disease 2 5<br />

Response rate (95%<br />

confidence interval)<br />

26 65 (56–80)


Table 3 : Relation between biological markers and Response<br />

Her 2 neu<br />

positive<br />

Her 2 neu negative<br />

Pathological CR 2 4 6<br />

Non Pathological<br />

CR<br />

15 19 34<br />

Total 17 23 40<br />

P value 0.6 (Non Significant)<br />

Estrogen Receptor Estrogen Receptor<br />

positive<br />

negative<br />

Pathological CR 3 3 6<br />

Non Pathological<br />

CR<br />

23 11 34<br />

Total 26 14 40<br />

P value 0.4 (Non Significant)<br />

Her 2 neu<br />

positive<br />

Her 2 neu negative<br />

Clinical Response 12 14 26<br />

Non responders 5 9 14<br />

Total 17 23 40<br />

P value 0.52 (Non Significant)<br />

Estrogen Receptor Estrogen Receptor<br />

positive<br />

negative<br />

Clinical Response 15 11 26<br />

Non responders 9 3 16<br />

Total 26 14 40<br />

P value 0.33 (Non Significant)<br />

Table 4. Hematological and nonhematological adverse events (n = 40)<br />

Adverse event Grade 1 or 2<br />

No. <strong>of</strong> patients (%)<br />

Hematological<br />

Anemia<br />

Leucopenia<br />

Neutropenia<br />

Thrombocytopenia<br />

Nonhematological<br />

Alopecia<br />

Anxiety<br />

Asthenia<br />

Conjunctivitis<br />

Cutaneous<br />

Diarrhea<br />

Hypersensitivity<br />

Infection<br />

Nail disorder<br />

Nausea/Vomiting<br />

Paresthesia<br />

Peripheral edema<br />

Stomatitis<br />

2 (5)<br />

20 (50)<br />

20 (50)<br />

4 (10)<br />

40 (100)<br />

0<br />

24 (60)<br />

10 (25)<br />

20 (50)<br />

16 (40)<br />

4 (10)<br />

6 (15)<br />

22 (55)<br />

16 (40)<br />

21 (42.5)<br />

9 (22.5)<br />

14 (35)<br />

Grade 3 or 4<br />

No. <strong>of</strong> patients (%)<br />

1 (2.5)<br />

0<br />

2 (5)<br />

0<br />

1 (2.5)<br />

8 (20)<br />

2 (5)<br />

6 (15)<br />

2 (5)<br />

0<br />

1 (2.5)<br />

7 (17.5)<br />

0<br />

2 (5)<br />

0<br />

2 (5)<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 23


original article <<br />

The Role <strong>of</strong> Pathologist in Modern <strong>Oncology</strong> Practice<br />

Hanaa Bamefleh, MBchB, FRCPC 1 , Abdul-Rahman Jazieh, MD, MPH 2<br />

(1) Department <strong>of</strong> Pathology and Laboratory Medicine<br />

(2) Department <strong>of</strong> <strong>Oncology</strong><br />

King Abdulaziz <strong>Medical</strong> City, Riyadh, KSA<br />

King Saud bin Adbulaziz University for Health Sciences, Riyadh, KSA<br />

Corresponding Author: Abdul-Rahman Jazieh, MD, MPH<br />

Chairman, Department <strong>of</strong> <strong>Oncology</strong> (Mail code 1777)<br />

King Abdulaziz <strong>Medical</strong> City, Kingdom <strong>of</strong> Saudi <strong>Arab</strong>ia<br />

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

Keywords: Pathologist, <strong>Oncology</strong>, Cancer Diagnosis, Tumor Boards<br />

ISSN: 2070-254X<br />

Abstract<br />

Background: Pathologists are <strong>of</strong>ten viewed as the invisible members <strong>of</strong><br />

oncology multidisciplinary teams, yet major decisions about patient care are<br />

totally dependent on their input. Making an accurate diagnosis is the first and<br />

most crucial step in managing cancer patients, which is the pathologist’s arena.<br />

Methods: Information about the topic was compiled from literature review,<br />

pr<strong>of</strong>essional societies publications, colleagues input and personal experience.<br />

Results: The purpose <strong>of</strong> this manuscript is to illustrate and outline the<br />

comprehensive role that should be fulfilled by a competent pathologist in<br />

providing state <strong>of</strong> the art service to oncology practices and to encourage<br />

maximum utilization <strong>of</strong> this role. The pathologist’s role in oncology practices is<br />

becoming increasingly critical due to the accelerated rate <strong>of</strong> new diagnostic tools<br />

and tests used in clinical applications. The roles <strong>of</strong> pathologist’s include making<br />

accurate diagnoses, evaluating disease response, participating in tumor boards,<br />

educating staff and conducting research.<br />

Conclusion: The role <strong>of</strong> pathologist is central to modern oncology practice and<br />

the pathologist is an integral member <strong>of</strong> the oncology care team.<br />

Various medical specialties and subspecialties have emerged over the last few<br />

decades creating increasing complexities in functions and roles <strong>of</strong> various heath<br />

care pr<strong>of</strong>essionals. To ensure best patient care, a strong cooperation among these<br />

subspecialties is crucial.<br />

Some specialties are essential for any tertiary health care center such as laboratory<br />

medicine and radiology, because all other services are dependent on them. For<br />

example, a surgeon may need the guidance <strong>of</strong> the pathologist preoperatively and<br />

intraoperatively for optimal completion <strong>of</strong> surgery. Similarly, an oncologist will<br />

not be able to treat a cancer patient appropriately without a proper diagnosis<br />

made by a pathologist.<br />

Since many <strong>of</strong> their activities are done away from the clinic or the patients,<br />

pathologists are considered by some as invisible members <strong>of</strong> oncology care team<br />

in spite <strong>of</strong> their critical role.<br />

The pathologist has multiple roles in the contemporary and future oncology<br />

practice that are presented and discussed in this manuscript. (Table 1)<br />

Making an accurate diagnosis<br />

To ensure that a pathologist carries this task effectively and efficiently to<br />

generate accurate specific diagnostic reports, he/she has to be updated on all<br />

issues relevant to tissue handling, evaluation and interpretation. The College <strong>of</strong><br />

American Pathologists (CAP) <strong>of</strong>fers anatomic pathology educational programs<br />

that are designed to improve the analytic and diagnostic skills <strong>of</strong> pathologists<br />

and technologists. 1 These skills and techniques have to be translated into a clear,<br />

accurate, surgical pathology report that indicates the diagnosis. 2 The Pathologists<br />

should know their limitations and capabilities and they should ask for help when<br />

needed. Inaccurate interpretation <strong>of</strong> pathology samples in any given patient may<br />

lead to providing erroneous management which must be avoided at all costs.<br />

Helping clinicians interpret the laboratory data to synthesize a clearer<br />

clinical picture<br />

How was the diagnosis reached should be highlighted by explaining all specific<br />

studies used to reach it, such as special stains, immunohistochemistry, molecular<br />

studies, electron microscopy and flowcytometry.<br />

How these studies correlate with the behavior or prognosis <strong>of</strong> the tumor has to<br />

be stated in the report.<br />

Correlation with any previous tissue or cytology samples taken from the patient<br />

has to be made to explain clearly whether they are the same pathology or two<br />

different pathologies. Furthermore, the updated pathologic staging, grading<br />

and status <strong>of</strong> resection margins are all essential information for any report<br />

on malignant lesion. A standardized reporting format has to be followed.<br />

References are available in many sites such as CAP or reference books. 3,4<br />

The need for more tissue should be mentioned in the report with explanation<br />

why it is needed. Is it for more studies essential for diagnosis, prognosis and<br />

management or because the tumor is very heterogeneous and different areas <strong>of</strong><br />

the tumor may show different morphology such as in many sarcomas, or just<br />

simply because the first biopsy was insufficient. Another scenario for requiring<br />

more tissue is the presence <strong>of</strong> carcinoma insitu in a superficial biopsy, where the<br />

possibility <strong>of</strong> invasion cannot be ruled out and a deeper biopsy is needed. The<br />

pathologist opinion will influence the sequence <strong>of</strong> the patient workup and enable<br />

24 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


the clinician to select the best diagnostic procedure. Adequate tissue specimen is<br />

essential for securing a final and accurate diagnosis.<br />

The pathologist should be receptive for the treating physician input regarding<br />

any missing relevant data from the report thus monitoring completeness <strong>of</strong> the<br />

surgical pathology report.<br />

All available data has to be reported using clear, precise words, avoiding vague,<br />

equivocal, or ambiguous terminology; keeping in mind that every word may be<br />

utilized by the oncologist / clinician to tailor the treatment plan for the patient.<br />

Assessing response to treatment<br />

In cases <strong>of</strong> patients with solid tumors who have received neoadjuvant therapy and<br />

in those with hematologic malignancies who have received induction therapy,<br />

the pathologist should comment on presence, grade and percentage <strong>of</strong> viable<br />

tumor, if it exists. Furthermore, comparison <strong>of</strong> tumor grade and differentiation<br />

in the current tissue sample with the previous samples is very important to plan<br />

further management.<br />

Evaluating and assessing new diagnostic technology application and<br />

utilization in clinical practice<br />

The pathologist should evaluate all new technologies in the field and decide with<br />

the clinicians about acquiring it to their practice.<br />

The behavior and prognosis <strong>of</strong> many cancers are better elucidated by the<br />

recent advanced in the genomic and molecular field. Pathologists should not<br />

only specify tumor size, lymph node status but also should state expression <strong>of</strong><br />

certain markers with prognostic or predictive values. Pathologists have become<br />

central in the development, validation, implementation and appropriate use <strong>of</strong><br />

predictive testing to better treat patients with targeted therapy. 5 Examples <strong>of</strong> such<br />

targets include over expression and or amplification <strong>of</strong> HE-2 Neu Oncogen, the<br />

expression or activation <strong>of</strong> epidermal growth factor receptor – Tyrosine Kinase<br />

pathway and over expression <strong>of</strong> C-Kit in gastrointestinal stromal tumor and<br />

leukemia. 6 Regular update <strong>of</strong> the pathology department policy and procedure<br />

manual for the proper and state-<strong>of</strong>- the-art way <strong>of</strong> handling and reporting <strong>of</strong> each<br />

tissue type should be done and maintained in the department. The oncology<br />

clinical team members should be kept abreast about all these updates.<br />

By providing this diagnostic information and by characterizing the biologic<br />

behavior <strong>of</strong> the tumor, the pathologist plays a critical role in the oncology team<br />

and impacts tremendously the patient’s care.<br />

Role in multidisciplinary teams and tumor boards<br />

The contributions <strong>of</strong> pathologists in the multidisciplinary meetings and tumor<br />

boards are essential. The pathologist does not just present slides, discuss<br />

differential diagnosis and suggest further work up; but he/she also answers<br />

queries, explains reasons for incomplete report and reasons for requesting more<br />

tissue or second opinion.<br />

This integrated care through the multidisciplinary approach has been widely<br />

recognized by different specialties. A good example is the many multidisciplinary<br />

breast teams that have been established all over the world. 5<br />

Education<br />

The pathologist should contribute and participate in the education <strong>of</strong> the clinical<br />

care teams whether in form <strong>of</strong> didactic lectures, tumor boards, or individual<br />

encounters to discuss specific cases and update the group on the most recent<br />

advances in the field.<br />

Effective communication<br />

there are many ways to deliver the report to the clinician. However, direct<br />

communication is essential to clarify any ambiguous issues and to get critical<br />

points through. For cases with insufficient clinical information or ambiguous<br />

findings, additional information should be sought out. Some cases may require<br />

urgent intervention and therefore, personal communication will assure timely<br />

action. Documentation <strong>of</strong> this discussion has to appear in the final report, which<br />

can be sent via intranet to patient’s electronic file and to responsible physicians<br />

by email as well as by sending a hard copy to patients file or doctors <strong>of</strong>fice. 7,8<br />

In a survey done by Zarbo, the greatest opportunities for improvement <strong>of</strong> anatomic<br />

pathology service were determined to relate not to pr<strong>of</strong>essional competence<br />

but to communication. 9 These include timeliness <strong>of</strong> reporting, communication<br />

<strong>of</strong> relevant information and the mandated notification <strong>of</strong> significant abnormal<br />

results. i.e critical value which requires immediate contact <strong>of</strong> the physician to<br />

rapidly initiate/modify treatment or to perform further required timely evaluation<br />

(Table 2 & 3). However, since there are no established critical value guidelines<br />

in surgical pathology, some situations may arise that require a common sense<br />

and personal experience <strong>of</strong> the pathologist to determine when an immediate<br />

communication with the clinician is needed. 8<br />

When communicating verbally with team members <strong>of</strong> the tumor board,<br />

pathologist should clearly explain all the terminology and descriptions used in the<br />

report and should correct any formatting problems to the reports during interface<br />

that might change the meanings. 2 This step is important to avoid misconstruction<br />

that can happen due to unawareness <strong>of</strong> some physicians to certain terminology<br />

used in surgical pathology practice. For example, some physicians think that<br />

metaplasia and dysplasia are a premalignant condition and the terms can be used<br />

interchangeably; while it is true for the later but not for the former term unless it<br />

becomes an atypical metaplasia.<br />

It is essential to deliver the diagnosis <strong>of</strong> unexpected malignancies, incomplete<br />

surgical resections, positive margins, unpredicted type <strong>of</strong> malignancy, frozen<br />

section results, discrepancies between frozen section and permanent section<br />

immediately and clearly to the clinicians, as these are critical findings that<br />

impact the management decisions. 7,8<br />

Any change <strong>of</strong> the final diagnosis for any reason such as obtaining more clinical<br />

information, getting more stains, more levels <strong>of</strong> the tissue blocks, more tissue<br />

from the same lesion should be delivered immediately to the oncologist and<br />

documented in the final amended report. The quality control <strong>of</strong>ficer <strong>of</strong> the<br />

laboratory should be informed and the reason for changing the report should be<br />

documented.<br />

<strong>Oncology</strong> research<br />

Well-structured research projects can be generated utilizing the data provided<br />

by the pathologists in the form <strong>of</strong> elaborate standardized reports that are well<br />

archived and easily retrievable. Correlative studies can be conducted for newly<br />

discovered tumor markers, molecular findings, and genetic alteration or <strong>of</strong> the<br />

different tumor types with response to different treatment modalities, behavior<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 25


original article <<br />

and prognosis. By putting together all these pathological data with the clinical<br />

details, brilliant research projects can be performed that will direct future<br />

diagnostic or management approaches and help more patients.<br />

The breakthroughs in molecular oncology led the pathologist to play a key-role<br />

in translational research for the identification <strong>of</strong> new targets in tissue specimens<br />

that may eventually lead to new therapeutics. 10<br />

Conclusion<br />

The pathologist roles in the modern oncology practice are complex and diverse<br />

but they are essential and critical for patient care, medical staff education and the<br />

advancement <strong>of</strong> medical knowledge and research.<br />

Acknowledgement<br />

The authors are indebted to Dr. Walid Khalbus for his critical review <strong>of</strong> this<br />

manuscript.<br />

References<br />

1. www.cap.org<br />

2. Valenstein PN. Formatting pathology reports. Applying four design<br />

principles to improve communication and patient safety. Arch pathol lab<br />

med 2008; 132(2):206-10.<br />

3. Rosai & Ackerman’s surgical pathology, Juan Rosai Mosby, Ninth Edition,<br />

2004.<br />

4. Susan C. Lester. Manual <strong>of</strong> surgical pathology, Susan C. Lester. Elsevier<br />

Churchil Livingstone, second edition, 2006.<br />

5. Masood S. The Expanding Role <strong>of</strong> Pathologists in the Diagnosis and<br />

Management <strong>of</strong> Breast cancer: worldwide Excellence in Breast Pathology<br />

Program. The Breast <strong>Journal</strong> 2003;9(2);s94-s97.<br />

6. Hartmann A. Role <strong>of</strong> predictive pathology in oncology-example <strong>of</strong> new<br />

therapies targeting EGFR. Verh Dtsch GCS Pathol 2006; 90:128 – 35.<br />

7. LiVolsi VA. Critical Values in Anatomic Pathology, How Do We<br />

Communicate? Am J Clin Pathol 2004; 122:171-172.<br />

8. Pereira TC, Liu Y, and Jan Silverman JF. Critical values in surgical<br />

pathology. Am J Clin Pathol 2004;122: 201-205.<br />

9. Zarbo RJ. Determining Customer Satisfaction in Anatomic Pathology.<br />

Arch Pathol Lab Med 2006; 130 (5): 645-649.<br />

10. Spatz A, Ruiter DJ, Busch C, Theodorovic I, Oosterhuis J.W, on behalf<br />

<strong>of</strong> the EORTC pathology group; The role <strong>of</strong> the EORTC pathologist in<br />

clinical trials: achievements and prospectives. European <strong>Journal</strong> <strong>of</strong> Cancer<br />

2002;28:S120-S24.<br />

Table 1: The Roles <strong>of</strong> the Pathologists in <strong>Oncology</strong> Practice<br />

1. Making an accurate diagnosis using all available tools including<br />

conventional and state-<strong>of</strong>-the-art methods<br />

2. Helping clinicians interpret the laboratory data to synthesize a clearer<br />

clinical picture.<br />

3. Helping in assessing and determining disease response.<br />

4. Evaluating and assessing new diagnostic technology application and<br />

utilization in clinical practice.<br />

5. Playing an active role in the multidisciplinary teams and tumor boards.<br />

6. Participating in the staff education.<br />

7. Maintaining communication with clinicians to provide critical<br />

information in a timely fashion.<br />

8. Participating actively in oncology research.<br />

Table 2: Critical values related to oncology patient<br />

1. Incomplete surgical resection<br />

2. Positive margins (margin involved by tumor)<br />

3. Discrepancy between frozen section & permanent section<br />

4. Change <strong>of</strong> a benign diagnosis to malignant<br />

5. Recurrence <strong>of</strong> tumor<br />

6. Unexpected malignancy (see table 3)<br />

7. Unpredicted type/grade or stage <strong>of</strong> malignancy<br />

8. Negative resection specimen<br />

9. Malignancy in superior vena cava syndrome<br />

10. Major change <strong>of</strong> final diagnosis for any reason<br />

11. Drug toxicity (e.g. lung, kidney, skin reaction)<br />

12. Organisms due to immuno suppression e.g. Mycobacterium<br />

tuberculosis, fungus, invasive aspergillosis, virus<br />

13. Neoplasm causing paralysis<br />

14. Large vessel in a core biopsy<br />

Table 3: Examples <strong>of</strong> unexpected malignancies<br />

1. Lymphoma in a gastric biopsy for Helicobacter<br />

2. Molar pregnancy in routine dilatation and curettage<br />

3. Lymphoma in fine needle aspiration <strong>of</strong> a lymph node suspected as<br />

tuberculosis<br />

4. Benign tumor with a focus <strong>of</strong> malignancy [table 4]<br />

5. Hematologic malignant neoplasms AML type M3, Burkitt’s lymphoma,<br />

and leukemia cutis. 8<br />

1.<br />

Table 4: Examples <strong>of</strong> Benign lesion /tumor with an unexpected<br />

focus <strong>of</strong> malignancy<br />

Carcinoma ex-pleomorphic adenoma<br />

2. Fibroadenoma with a focus <strong>of</strong> ductal carcinoma<br />

3. Mature cystic teratoma containing immature elements or carcinoma<br />

4. Benign prostatic hypertrophy with focal prostatic carcinoma<br />

5. Nodular goiter with microscopic focus <strong>of</strong> papillary carcinoma.<br />

6. Undescended tests with intratubular germ cell neoplasia and /or germ<br />

cell tumor.<br />

7. Umbilical hernia with metastatic adenocarcinoma<br />

26 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


notes <<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 27


original article <<br />

Taxanes Versus Vinorelbine and 5-Fluorourocil as a First Therapy in Metastatic<br />

Breast Cancer<br />

Ibtessam Saad El-Din, MD 1 , Wael Samir, MD 1 , Wessam El-Sherief, MD 1 , Yomna Khaiery, M.Sc 2<br />

(1) Kasr El-Aini <strong>Oncology</strong> Center (NEMROCK)<br />

(2) El-Salam <strong>Oncology</strong> Center<br />

Corresponding Author: Wessam Ali El-Sherief, MD<br />

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

Keywords: Metastatic Breast Cancer, Taxanes, Vinorelbine and 5-FU<br />

ISSN: 2070-254X<br />

Abstract<br />

Purpose: To compare the efficacy and tolerance <strong>of</strong> taxanes versus combination<br />

<strong>of</strong> vinorelbine and 5-FU in the treatment <strong>of</strong> MBC previously treated with<br />

anthracyclines and analyzing the effect <strong>of</strong> various prognostic factors.<br />

Material and methods: Between April 2005 and April 2007, sixty eligible<br />

MBC patients were randomized between 2 arms. Arm I received taxanes (either<br />

paclitaxel 175 mg/m2 D or Docetaxel 100 mg/m 1 2 D ) and arm II received<br />

1<br />

combination <strong>of</strong> vinorelbine 25 mg/m2 D and D , and 5- FU 600 mg/m 1 5 2 by<br />

continuous IV infusion D1 to D5. cycles <strong>of</strong> chemotherapy were repeated every<br />

21 to 28 days for a total <strong>of</strong> 6 cycles in both arms.<br />

Results: Three patients (10.3%) in arm I achieved complete remission versus<br />

one patient (3.4%) in arm I, while 13 patients (44.8%) in arm I versus 9 patients<br />

(31%) in arm II had partial remission. (P=0.383). Neurosensory changes and<br />

fluid retention were more evident is arm I while diarrhea and nail changes were<br />

more common in arm II (P=0.03). The mean overall survival was 27.7 months<br />

with taxanes and 26.5 months with vinorelbine and 5-FU (P = 0.79) while<br />

progression free survival was 15.7 months in patients treated in arm I versus 8.5<br />

months in arm II (P = 0.234).<br />

Conclusion: Both regimens are well tolerated and feasible, there treatment<br />

outcome and toxicity pr<strong>of</strong>ile are comparable but cost effective issue may have an<br />

important impact on treatment selection.<br />

Introduction<br />

Breast cancer continuous to be the most common cancer in women world-wide.<br />

Metastatic breast cancer is usually considered as an incurable situation for which<br />

treatment chosen to control the disease should take into account the maintenance<br />

<strong>of</strong> a good quality <strong>of</strong> life. The clinical benefit which encompasses objective<br />

response and long standing stability <strong>of</strong> the disease has <strong>of</strong>ten become a goal in<br />

metastatic setting. (1) Also, improvement in time to progression and duration <strong>of</strong><br />

response have been considered as primary goals <strong>of</strong> treatment. (2)<br />

Eventually, chemotherapy is considered for almost all patients with metastatic<br />

breast cancer. Several agents are now available <strong>of</strong>fering expanded treatment<br />

options for many patients. (3)<br />

Combination regimens should be considered for patients with overwhelming<br />

symptoms or rapidly progressive or life threatening metastasis for which a<br />

combination regimen is more likely to result in a tumor response. (4)<br />

Response rates to initial therapy with anthracyclines, taxanes, capecitabine,<br />

vinorelbine, and gemecitaline range on average from 25% to 60% with median<br />

time to progression averaging approximately 6 months. (4)<br />

The aim <strong>of</strong> this study is to compare the efficacy and tolerance <strong>of</strong> taxanes versus<br />

combination <strong>of</strong> intravenous vinorelbine and 5-FU given by continuous infusion<br />

in the treatment <strong>of</strong> metastatic breast cancer patients previously treated with<br />

anthracyclines and analyzing the effect <strong>of</strong> various breast prognostic factors in<br />

both regimens.<br />

Patients and Methods<br />

This study is a randomized prospective study that included 60 patients<br />

with metastatic breast cancer presented to Kasr El-Aini <strong>Oncology</strong> Center<br />

(NEMROCK) and El-Salam <strong>Oncology</strong> Center in the period from April 2005 to<br />

April 2007.<br />

Patient eligibility criteria:<br />

1. Patients age range from 18 to 70 years.<br />

2. WHO performance status <strong>of</strong> 0 to 2.<br />

3. Pathologically proved breast cancer with evidence <strong>of</strong> visceral, bone or<br />

locoregional metastasis.<br />

4. All patients should have been treated previously with an anthracycline as<br />

an adjuvant setting.<br />

Patients were randomized between two arms. Arm I received taxanes (either<br />

paclitaxel or docetaxel), and arm II received vinorelbine and 5-Flurouracil.<br />

Study design and treatment:<br />

Thirty patients were treated in arm I by a single agent, taxanes either docetaxal<br />

100 mg/m2 D 1 hour intravenous infusion or paclitxel 175 mg/m 1 2 D 3 hours<br />

1<br />

intravenous infusion, to be repeated from 3 to 4 weeks. Patients were given<br />

antiemetics, antihistaminecs and corticosteroids as premedications before<br />

receiving the taxane and continued two days after.<br />

28 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


Arm II included also 30 patients treated by a combination chemotherapy protocol<br />

consisting <strong>of</strong> vinorelbine (Navelbine) given intravenously in a dose <strong>of</strong> 25 mg/<br />

m 2 D 1 and D5 together with 5-Fluorouracil 600 mg/m 2 by a continuous infusion<br />

from D1 to D5 <strong>of</strong> the cycle, cycles to be repeated 21 to 28 days provided clinical<br />

and hematological recovery.<br />

Actual body weight was used to calculate body surface area, and after an<br />

amendment to the study protocol the maximum body surface was limited to 2.0<br />

m 2 . Clinical, hematological and biochemical assessments were required before<br />

each cycle, including assessment <strong>of</strong> toxic effects according to the common<br />

toxicity criteria, version 1 <strong>of</strong> the National Cancer Institute.<br />

In both arms patients were re-evaluated by physical examination, radiological<br />

imaging and laboratory tests after completion <strong>of</strong> 3 cycles <strong>of</strong> the chemotherapy<br />

protocol to assess the response as well as the efficacy and toxicity <strong>of</strong> the<br />

treatment.<br />

Patients showing disease progression were planned to shift to another line <strong>of</strong><br />

chemotherapy while those showing response or even stable disease continued on<br />

the same line <strong>of</strong> treatment to a total <strong>of</strong> 6 cycles in both arms <strong>of</strong> the study.<br />

Statistical Analysis<br />

Data were statistically described in terms <strong>of</strong> range, mean, standard deviation<br />

(± SD), median, frequencies (number <strong>of</strong> cases) and relative frequencies<br />

(percentages) when appropriate. Comparison <strong>of</strong> age between the study groups<br />

was done using Mann Whitney T-test for independent samples for comparing<br />

categorical data, Chi square (x2) test was; performed. Yates correction was<br />

used instead when the frequency is less than 10. Survival, analysis was done<br />

for the different outcome measures using Kaplan Mayer statistics with the<br />

corresponding survival graphs, A probability value (P-value) less than 0.05 was<br />

considered, statistically significant. All statistical calculations were done using<br />

computer programs. Micros<strong>of</strong>t Excel version 7 (Micros<strong>of</strong>t Corporation, NY,<br />

USA) and SPSS (Statistical Package for the Social Science; SPSS Inc., Chicago,<br />

IL, USA) statistical program.<br />

Results<br />

Patients characteristics:<br />

The characteristics <strong>of</strong> the 60 patients enrolled in the study are summarized in<br />

Table (1).<br />

Symptoms<br />

A variety <strong>of</strong> presenting symptoms were existing according to the different<br />

metastatic sites.<br />

The cardinal symptom was pain either in the right hypochondrium related to<br />

the liver metastasis <strong>of</strong> patients 9/30 (30%) in arm I and 5/30 (16.7%) in arm II<br />

(P- 0.22) or in bony metastatic sites 8/30(26.7%) in arm I and 12/30(40%) in<br />

arm II (P= 0.206).<br />

Chest symptoms (dyspnea, cough + expectoration) were found in 36.7 % (11/30)<br />

<strong>of</strong> patients in arm I and (30%) 9/30 in arm II (P = 0.392)<br />

Swelling (in the breast or lymph nodes) is 3/30 (10 %) <strong>of</strong> patients in arm I and<br />

4/30 (13.3%) in arm II (p-value0.5). Skin nodules in 12/30 (40%) <strong>of</strong> patients in<br />

arm I and 11/30(36.7) in arm II (P = 0.792).<br />

Number <strong>of</strong> metastasis:<br />

Number <strong>of</strong> metastatic sites in both arms are summerised in Table (2).<br />

Treatment related toxicity:<br />

Treatment related toxicities were measured from grade 0-4 according to the<br />

WHO recommendations as regard hematological toxicities (Table 3) and<br />

according to the EORTC classification and recommendation for grading <strong>of</strong> nonhematological<br />

toxicities (Table 4).<br />

Response:<br />

The assessment <strong>of</strong> response was carried out at the end <strong>of</strong> the treatment course.<br />

Patients who achieved complete response (CR) were 3/30 (10.3%) in arm I<br />

(received taxanes), versus 1/30 (3.4%) in arm II (received navelbine and 5-FU).<br />

Thirteen patients (44.8%) in arm I had partial remission (PR) versus 9 patients<br />

(31%) in arm II.<br />

While patients who developed disease progression were 3 patients (10.3%) in<br />

arm I versus 6 patients (31%) in arm II.<br />

Patients who had stationary disease (SD) were 11/30 (36.6%) in arm I versus<br />

14/30 (46%).<br />

The results <strong>of</strong> the response rate were statistically non significant between the two<br />

arms (P = 0.383).<br />

Survival:<br />

The mean overall survival (OAS) was 27.7 months in arm I versus 26.5 months<br />

in arm II (P = 0.79) (Fig. 1), while the progression free survival (PFS) for arm I<br />

was 15.7 months and 8.5 months for arm I (P = 0.234) (Fig. 2).<br />

Factors affecting response rate:<br />

Using umivariant analysis <strong>of</strong> different prognostic factors were studied in relation<br />

to response, time to progression and overall survival.<br />

Factors as age (>35 years), site <strong>of</strong> recurrence (local or bone) and number <strong>of</strong><br />

metastatic sites (


original article <<br />

Inspite adequate primary therapy in breast cancer, many patients with apparently<br />

localized disease harbor subclinical micrometastasis micrometastasis that may<br />

grow into clinically relevant macrometastases later on. (5)<br />

Metastatic breast cancer patients have a median survival <strong>of</strong> 2-3 years, this occurs<br />

despite the discovery <strong>of</strong> numerous new agents that can palliate the disease and<br />

more rarely increase the overall survival. (6) Therefore, for the majority <strong>of</strong> patients<br />

with metastatic breast cancer “cure” is not the goal <strong>of</strong> treatment ,instead, more<br />

conservative treatments are prefered to obtain maximum control <strong>of</strong> symptoms,<br />

prevent serious complications and prolong life with minimal toxicities and<br />

disruption <strong>of</strong> quality <strong>of</strong> life. (5)<br />

In the last decade the development <strong>of</strong> new cytotoxic drugs and combinations and<br />

the introduction <strong>of</strong> novel targeted agents have permitted to lenghthen patient’s<br />

survival and improve quality <strong>of</strong> life. (7)<br />

There is no single standard chemotherapy regimen for patients with MBC.<br />

In addition to the taxanes, various agents (e.g. capecitabine, gemcitabine and<br />

vinorelbine) used alone or in combination have emerged to be efficacious.<br />

In this retrospective study, we compared taxanes verus a combination <strong>of</strong><br />

vinorelbine & fluorouracil in patients with metastatic cancer breast. Complete<br />

remission CR was 3/30 (10.3%) in arm I (received taxanes) vs 1/30 (3.4%) in<br />

arm II (received navelbine &5FU) with no statisticaly significant difference.<br />

However difference in partial response and progressive disease was slightly<br />

better in Taxane group with no statisticaly significant difference.<br />

Patient who attained partial remission PR was 13/30(44.8%) in arm I vs 9/30<br />

(31%) in arm II, while patient who developed progressive disease PD was 3/30<br />

(10.3%) in arm I vs 6/30 (20.0%) in arm II. The mean over all survival was 27.7<br />

months vs 26.5 months in arm II.<br />

Factors affecting response were age, site <strong>of</strong> recurrence, number <strong>of</strong> metastasis all<br />

theses factors show statistically significant difference in both treatment groups,<br />

while other factors showed no statistically significant difference.<br />

These factors are performance status PS, menopausal status (MS), hormonal<br />

status (HS) as will as disease free survival (DPS). This response rate is<br />

comparaple to other study that use the same regimen like Bonneteterre et al. (8)<br />

who obtained response in 176 women with metastatic breast cancer who had<br />

failed anthracycline-based therapy The CR rate following Taxotere was 7%,<br />

compared to 4.4% following 5-FU and Navelbine. The PR rate was 36% for the<br />

Taxotere group and 34.4% for the combination group. Overall survival was 16<br />

months for the Taxotere group and 15 months for the combination group.<br />

Also in comparesion to other regimens used in MBC as vinorelbine and cispltin<br />

that reported by Vassilomanlakis et al. (9) , who obtaind response rate in 53<br />

patients with 4 patients (8%) in CR, 22 patients (41%) in PR, ORR (49%). In<br />

comparesion to Gunel et al. (10) who obtaind response rate25% in 24 patient and<br />

to Ray-Coquard et al. (1998) who obtaind response rate in 58 patients 2 patients<br />

(3%) in CR,22 patients (38%), ORR (43%) used the same regimen.<br />

There is wide range <strong>of</strong> variation between different studies using Chemotherapy<br />

in MBC even between studies using the same regimen, this might be due to<br />

selection bias where these studies are mostly non-randomized trial.<br />

Other important factor is the use <strong>of</strong> different dose schedules among different<br />

studies, not only the scheduled dose differ from study to study but more<br />

important the relative average dose intensity differ from study to study even if<br />

they have got same dose schedule.<br />

The number <strong>of</strong> patients needed hospitalization with blood transfusion and<br />

growth stimulating factor were 4 vs 5 for taxanes vs navelbine & 5-FU average<br />

hospital stay is four days, these adds to treatment cost.<br />

In comparison to Bonneterre et al. (8) who repoted the main grade III-IV toxicities<br />

were 82% vs 67%; stomatitis 5% vs 40%; febrile neutopenia 13%vs 22% and<br />

infection 2% vs 7% for docetaxel vs navelbine & 5-FU. There was one possible<br />

treatment-related death in docetaxel arm and five with navelbine & 5-FU.<br />

As regards cost effective outcome, the paclitaxel or docetaxel arm costs about<br />

35,000 L.E. and the navelbine &5-FU arm costs about 15,000 L.E. and this<br />

reflects the economical advantage <strong>of</strong> the vinorelbine & 5-flurouoracil.<br />

This economic advantage considered as a major advantage especially in favor <strong>of</strong><br />

navelbine & 5-FU in developing countries.<br />

Clinical research in which more attention is paid to symptom control and quality<br />

<strong>of</strong> life and increased emphasis is placed on improving treatment individualization<br />

remains the only way for progress to be made in the management <strong>of</strong> MBC<br />

In conclusion, in a situation where treatment outcome as well as toxicity pr<strong>of</strong>ile<br />

are comparable in both treatment arms, the cost effective issue should have an<br />

important impact on treatment selection. With the growing understanding <strong>of</strong><br />

biology <strong>of</strong> breast cancer and the advent <strong>of</strong> new techniques such as genomics<br />

and proteomics, multiple new targets for anticancer therapy are identified every<br />

year. The challenge for clinicians will be to find the most appropriate niche for<br />

the new biologic therapies in breast cancer management, the hope being that the<br />

therapies will significantly improve the quality and the length <strong>of</strong> chemotherapy<br />

induced remissions and disease stabilization.<br />

References<br />

1. Orlando AS: Arm Oncol, 2007; 18 suppl 16: vi 74-6.<br />

2. Cathie TC & Robert WC: Goals and Objectives in the Management <strong>of</strong><br />

Metastatic Breast Cancer. The Oncologist, December 2003; Vol. 8, No.6,<br />

514-520.<br />

3. Bernard-Marty, Fatima Cardoso, Martine J: Facts and Controversies in<br />

Systemic Treatment <strong>of</strong> Metastatic Breast Cancer The Oncologist, November<br />

2004; Vol. 9, No. 6, 617-632.<br />

4. Costanza ME, Weiss RB, Henderson IC: Safety and efficacy <strong>of</strong> using a<br />

single agent or a phase II agent before instituting standard combination<br />

chemotherapy in previously untreated metastatic breast cancer patients:<br />

report <strong>of</strong> a randomized study-Cancer and Leukemia Group B 8642. J Clin<br />

Oncol 1999; 17:1397.<br />

5. Colozza Mariantonie, Evandro De Azambuja, Nicola Personeni:<br />

Achievements in systemic therapies in the pregenomic era in MBC. The<br />

oncologist; March, 2007; 12, 253-270.<br />

6. Bontenbal M, Creemers GJ, Braun HJ: Phase II to III study comparing<br />

doxorubicin & docetaxel with 5-FU, doxorubicin & cyclophosphamide as first<br />

line chemotherapy in patients with MBCJ Clin Oncol, 2005; 23, 7081-7088.<br />

30 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


7. Adamo V, Franchina T, Adamo B, Ferraro G, Rossello R, Maugeri Sacca<br />

M, Scibilia C, Valerio MR & Russo A: Safety and activity <strong>of</strong> trastuzumab-<br />

containing therapies for the treatment <strong>of</strong> metastatic breast cancer: our long-<br />

term clinical experience (GOIM study) Annals <strong>of</strong> <strong>Oncology</strong> 18 (Supplement<br />

6): vill-vil5, 2007 doi: l0.10937 annonc/mdm217.<br />

8. Bonneterre J, Roche H, Monnier A: Docetaxel vs 5-fluorouracil plus<br />

vinorelbine in metastatic breast cancer after anthracycline therapy failure.<br />

British Journl <strong>of</strong> Cancer. 2002; 87:1210-1215.<br />

9. Vassilomanolakis M, Koumakis G, Barbounis V: First-line chemotherapy<br />

with docetaxel and cisplatin in metastatic breast cancer patients previously<br />

treated with anthracyclines.Ann oncol 2000; 11: 1155-1 1160.<br />

10. Gunel N, Akcail Z, Yamac D,Onuk E Yilmaz E, Bayram Tekin E, Coskun<br />

U et al.: Cisplatin plus vinorelbine as a salvage regimen in refractory breast<br />

cancer turners 2000 Jul -Aug; 86 (4) :283 -50.<br />

11. Ray-Coquard I, Biron P, Baachelot T et al.: Vinorelbine and cisplatin<br />

Figures<br />

(CIVIC regimen) for the treatment <strong>of</strong> metastatic breast carcinoma after<br />

failure <strong>of</strong> anthracycline and/or paclitaxl. co regimines 1998; 82: 134-40.<br />

Fig. 1: Overall survival<br />

Fig. 2: Progression free survival<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 31


original article <<br />

Tables<br />

Table 1: Patients characteristics<br />

Parameter<br />

Age in years < 35<br />

>35<br />

Performance status 0<br />

1<br />

2<br />

Menopausal status Pre<br />

Post<br />

Pathology IDC<br />

ILC<br />

Tumor grade II<br />

III<br />

ER group +ve<br />

-ve<br />

PR group +ve<br />

-ve<br />

Her 2neu +ve<br />

-ve<br />

Site <strong>of</strong> metastasis Visceral:<br />

Liver<br />

Lung<br />

Table 2: Number <strong>of</strong> metastatic sites in both arms<br />

Arm I Arm II<br />

Number Percent Number Percent<br />

5<br />

16.5%<br />

7<br />

23.3%<br />

25<br />

83%<br />

23<br />

76.6%<br />

5<br />

17.2%<br />

4<br />

14.8%<br />

18<br />

62.1%<br />

19<br />

66.7%<br />

6<br />

20.7%<br />

5<br />

18.5%<br />

10<br />

33.3%<br />

11<br />

37.9%<br />

20<br />

66.7%<br />

18<br />

62.1%<br />

26<br />

86.6%<br />

24<br />

80%<br />

4<br />

13.3%<br />

6<br />

19%<br />

20<br />

66.7%<br />

22<br />

73.3%<br />

10<br />

33.3%<br />

8<br />

26.7%<br />

18<br />

62.1%<br />

22<br />

73.3%<br />

11<br />

37.9%<br />

8<br />

26.7%<br />

17<br />

58.6%<br />

18<br />

60%<br />

12<br />

41.4%<br />

12<br />

40%<br />

3<br />

37.5%<br />

0<br />

0%<br />

5<br />

62.5%<br />

3<br />

100%<br />

12<br />

12<br />

40%<br />

40%<br />

32 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org<br />

8<br />

9<br />

26.7%<br />

30%<br />

P-value<br />

0.112<br />

0.581<br />

0.789<br />

0.551<br />

0.576<br />

0.412<br />

0.562<br />

0.339<br />

0.206<br />

0.294<br />

Loco 15 50% 14 46.7% 0.500<br />

Bone 9 30% 12 40% 0.294<br />

Number <strong>of</strong> metastatic sites<br />

No<br />

Arm I<br />

% No<br />

Arm II<br />

%<br />

One site 15 50 16 53.3<br />

Two sites 12 40 14 46.7<br />

Three sites 3 10 0 0<br />

Table 3: Hematological toxicities<br />

Parameter<br />

Neutropenia I,<br />

&III<br />

II<br />

Anaemia I,<br />

&III<br />

II<br />

Thrombo-cytopenia I,<br />

&III<br />

II<br />

P-value<br />

0.386<br />

No<br />

Arm I<br />

% No<br />

Arm II<br />

%<br />

P-value<br />

12<br />

40% 10<br />

33.3% 0.395<br />

0<br />

0<br />

4<br />

13.3% 3<br />

10% 0.690<br />

0<br />

0<br />

2<br />

6.7% 1<br />

3.3% 0.5<br />

0<br />

0


Table 4: Non-hematological toxicities<br />

Parameter Grade<br />

Number<br />

Arm I<br />

Percent<br />

Arm II<br />

Number Percent<br />

P-value<br />

Alopecia I ,II & III 6<br />

20% 2<br />

6.7% 0.12<br />

0<br />

0<br />

Fatigue I ,II & III 9<br />

30% 9<br />

30% 0.61<br />

0<br />

0<br />

Nausea I ,II & III 9<br />

30% 7<br />

23.3% 0.56<br />

0<br />

0<br />

Nail changes I ,II & III 0<br />

0% 4<br />

13.8% 0.03<br />

0<br />

0<br />

Fluid ret. > 3kg 8 26.7% 0 0 0.03<br />

Vomiting I ,II & III 7<br />

24.1% 12<br />

40% 0.196<br />

0<br />

0<br />

Diarrhea I ,II & III 0<br />

0 4<br />

13.3% 0.038<br />

0<br />

0<br />

Epiphora Positive 3 10% 2 6.9% 0.66<br />

Stomatitis I ,II & III 7<br />

23.3% 5<br />

16.7% 0.51<br />

0<br />

0<br />

Neurosens I ,II & III 7<br />

23.3% 1<br />

3.3% 0.02<br />

0<br />

0<br />

Neuromot I ,II & III 3<br />

10% 0<br />

0% 0.68<br />

0<br />

0<br />

Table 5<br />

Prognostic factor Response TTP OAS<br />

Age 0.014 0.034 0.001<br />

Site <strong>of</strong> recurrence 0.004 0.012 0.026<br />

No <strong>of</strong> metastatic sites 0.008 0.016 0.03<br />

Table 6<br />

Prognostic factor Response TTP OAS<br />

PS 0.794 0.727 0.317<br />

MS 0.544 0.118 0.119<br />

HS 0.452 0.969 0.668<br />

PS : Performance status MS: Menopausal status HS: Hormonal status<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 33


original article <<br />

Dose-Dense Chemotherapy in High-Risk Breast Cancer: Treatment Outcome and<br />

Toxicity<br />

Salah El-Mesidy, MD, Mohsen Mokhtar, MD, Amr El-Kashif, MD, Loay Kasem, M.Sc<br />

Department <strong>of</strong> Clinical <strong>Oncology</strong>, Faculty <strong>of</strong> Medicine, Cairo University.<br />

Corresponding Author: Dr. Amr El-Kashif, MD<br />

E-mail: amrelkashif@hotmail.com<br />

Keywords: Dose-Dense, Adjuvant Chemotherapy, Breast Cancer.<br />

ISSN: 2070-254X<br />

Abstract<br />

Purpose: To prospectively assess the treatment outcome <strong>of</strong> dose-dense adjuvant<br />

doxorubicin and cyclophosphamide (AC) followed by paclitaxel (T) in highrisk<br />

breast cancer patients (dose-dense arm) and to compare it with the available<br />

treatment results <strong>of</strong> the conventionally scheduled fluorouracil, doxorubicin,<br />

and cyclophosphamide (FAC) used to treat high-risk patients (control arm).<br />

Study endpoints included relapse-free survival (RFS), overall survival (OS) and<br />

toxicity.<br />

Patients and Methods: After mastectomy or breast conservative surgery,<br />

high-risk node-positive breast cancer 61 patients were assigned to receive<br />

adjuvant 4 cycles <strong>of</strong> doxorubicin/cyclophosphamide followed by 4 cycles <strong>of</strong><br />

paclitaxel (AC/T) every 2 weeks. The treatment outcome <strong>of</strong> dose-dense AC/T<br />

was eventually compared with that <strong>of</strong> the conventionally treated high-risk<br />

patients using adjuvant 6 cycles <strong>of</strong> FAC combination chemotherapy scheduled<br />

every 3 weeks. The relevant data <strong>of</strong> 43 patients received FAC chemotherapy<br />

were obtained from the medical records. Both arms were balanced regarding<br />

age, menopausal status, number <strong>of</strong> positive axillary lymph nodes and hormonal<br />

status.<br />

Results: At a median follow up <strong>of</strong> 37 months (range 12 – 48 months), the<br />

3-year adjusted RFS rates for AC-T and FAC were 76% and 54.6%, respectively<br />

(P=0.042) and the mean disease-free interval was 40.6 ± 7.2 months (95% CI,<br />

37.7 - 43.6) for dose-dense AC/T arm Vs 36.9 ± 6.9 months (95% CI, 32.3 -<br />

41.5) for FAC arm, (P=0.040). The subgroup analysis revealed that dose-dense<br />

chemotherapy had a statistically significant positive effect on the 3-year RFS in<br />

premenopausal patients, patients with 10 or more (N3) positive axillary lymph<br />

nodes, positive ER status. There was no statistically significant difference in the<br />

3-year OS between the two regimens. The dose-dense arm was associated with<br />

more grade 3-4 toxicity compared to FAC arm, mainly neuropathy (36%. Vs<br />

2.3%, P=0.002), anemia (49%. Vs 13.9%, P=0.037) and granulocytopenia (57%.<br />

Vs 213%, P=0.04). No toxicity-related mortality was observed in both arms.<br />

Conclusion: Dose-dense AC/T significantly improved the relapse-free survival<br />

in patients with high-risk primary breast cancer and was less well tolerated<br />

compared with the conventionally scheduled FAC. The benefit was evident in<br />

premenopausal patients, extensive axillary nodal metastasis and positive ER<br />

status.<br />

Introduction<br />

The value <strong>of</strong> adjuvant chemotherapy has been convincingly demonstrated in<br />

breast cancer patients with involved axillary nodes. In the last three decades,<br />

incremental benefits in breast cancer disease-free survival (DFS) and overall<br />

survival (OS) have occurred with the introduction <strong>of</strong> anthracycline and taxane<br />

adjuvant chemotherapy regimens and with the exploration <strong>of</strong> higher doses (dose<br />

intense) and more frequent administration <strong>of</strong> these drugs (dose dense) (1-3). The<br />

importance <strong>of</strong> dose-intensity for adjuvant chemotherapy in patients with breast<br />

cancer has first described by Hryniuk et al.(4). Higher dose-intensity can be<br />

achieved by either increasing the single dose per cycle (i.e. higher dose) or by<br />

reducing the intervals between cycles (i.e. dose density) (5).<br />

The concept called log-cell kill postulated that a given dose <strong>of</strong> an antineoplastic<br />

compound would always kill a certain fraction <strong>of</strong> the tumor, regardless <strong>of</strong> the<br />

number <strong>of</strong> cells present. On the basis <strong>of</strong> that model, higher doses are supposed<br />

to be effective because the fraction <strong>of</strong> destroyed cancer cells will increase with<br />

intensification <strong>of</strong> the dose. However, the impact <strong>of</strong> therapy is not only associated<br />

with the tumor-cell kill by each dose but also with the rate <strong>of</strong> cancer regrowth<br />

between cycles (6). Gompertzian kinetics suggest that micrometastases in the<br />

adjuvant setting grow faster than established macrometastases; thus, there is<br />

likely a higher regrowth <strong>of</strong> micrometastases between the cycles. Therefore, the<br />

administration <strong>of</strong> cytotoxic drugs with a shortened interval between treatments<br />

would be an even more effective strategy for minimizing residual tumor burden<br />

than pure dose escalation (5).<br />

In the National Cancer Institute <strong>of</strong> Canada (NCIC) Clinical Trials Group (CTG)<br />

MA5 trial, 6 months <strong>of</strong> cyclophosphamide, epirubicin, and fluorouracil (CEF)<br />

improved 5-year DFS and OS compared with 6 months <strong>of</strong> cyclophosphamide,<br />

methotrexate, and fluorouracil (CMF) (7). Subsequently, CEF was compared<br />

with a 12-week regimen <strong>of</strong> epirubicin and cyclophosphamide (EC) administered<br />

with higher doses and more frequent dosing in locally advanced breast cancer.<br />

No difference was detected between regimens in progression-free survival at 34<br />

months (8). Incorporating taxanes into anthracycline-based schedules yielded,<br />

in most studies, an additional benefit in both DFS and OS and dose-dense<br />

anthracycline-based and paclitaxel-based regimens have shown to be more<br />

effective than the conventional dosing schedule (9-13).<br />

34 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


The aim <strong>of</strong> the current study was to prospectively assess the treatment outcome<br />

<strong>of</strong> dose-dense adjuvant doxorubicin and cyclophosphamide (AC) followed <strong>of</strong><br />

paclitaxel (T) in high-risk breast cancer patients. Furthermore, the results <strong>of</strong> dosedense<br />

chemotherapy were compared with those <strong>of</strong> the conventionally scheduled<br />

combination chemotherapy <strong>of</strong> fluorouracil, doxorubicin, and cyclophosphamide<br />

(FAC) used to treat high-risk patients (control arm). Study endpoints included<br />

relapse-free survival (RFS), overall survival (OS) and toxicity.<br />

Patients and Methods<br />

The current prospective study included female patients with histologically<br />

proven node-positive high-risk invasive early breast cancer referred to Kasr<br />

El–Aini Center <strong>of</strong> Radiation <strong>Oncology</strong> and Nuclear Medicine (NEMROCK)<br />

during the period between January 2006 and December 2009. The study had the<br />

institutional ethical review board approval.<br />

Patient population: Patients eligible to receive dose-dense AC/T chemotherapy<br />

had the following criteria:<br />

• Age 18 - 65 years old.<br />

• Performance Status (PS) <strong>of</strong> 0–2 according to The Eastern Cooperative<br />

<strong>Oncology</strong> Group (ECOG) scale.<br />

• Operable breast cancer: T1-3, N1-2 treated by either mastectomy or<br />

breast conservative surgery (BCS) and axillary nodal dissection with free<br />

surgical margins.<br />

• High-risk features: either 4 or more positive axillary lymph nodes or 1-3<br />

positive nodes with over expression <strong>of</strong> the human epidermal growth factor<br />

receptor 2 (HER2/neu).<br />

• No evidence <strong>of</strong> metastatic disease.<br />

• No prior chemotherapy or radiotherapy received.<br />

• Adequate organ functions and No evidence <strong>of</strong> significant illness or comorbidities.<br />

Pre chemotherapy investigations included; 1- Laboratory investigations:<br />

complete blood picture, kidney function tests, liver function tests, CA15-3.<br />

2- Radiological assessment: baseline chest radiograph, abdominal ultrasound,<br />

echocardiography 3- Isotopic bone scanning. 4- Estrogen and progesterone<br />

receptors (ER and PR) status. 5- HER2/neu expression by immunohistochemistry:<br />

a score <strong>of</strong> (3+) was considered positive (overexpressed) while scores <strong>of</strong> (0, 1+,<br />

or 2+) were considered negative.<br />

Treatment regimen: Eligible patients were assigned to receive dose-dense AC/T<br />

adjuvant chemotherapy which consisted <strong>of</strong>: 4 cycles <strong>of</strong> AC (doxorubicin 60 mg/<br />

m2 and cyclophosphamide 600 mg/m2 administered IV on day 1) every 2 weeks<br />

followed by 4 cycles <strong>of</strong> T (paclitaxel 175 mg/m2 administered by IV infusion over<br />

3 hours on day 1) every 2 weeks. Prophylactic granulocyte colony-stimulating<br />

factor (G-CSF) was administered using filgrastim (5 ug/Kg/day subcutaneous<br />

injection) for 3 consecutive days 24-48 hours after chemotherapy. Treatmentinduced<br />

toxicity was evaluated according to the National Cancer Institute (NCI)<br />

Common Toxicity Criteria version 2.0 grading system (14). Patients who had an<br />

overexpressed HER2/neu did not receive adjuvant trastuzumab to exclude any<br />

positive effect on treatment outcome that may be attributed to its addition.<br />

After completion <strong>of</strong> chemotherapy, radiotherapy was given to the whole breast<br />

(after BCS) or chest wall ( after mastectomy) and peripheral lymphatics at a<br />

dose <strong>of</strong> 5040 cGy/28 fractions using linear accelerator 6 MV followed by a<br />

boost to the site <strong>of</strong> primary tumor(after BCS) at a dose <strong>of</strong> 1600-2000 cGy/8-10<br />

fractions by electron beam or photons. Premenopausal patients with ER and/or<br />

PR positive tumors were assigned to receive 5 years <strong>of</strong> adjuvant tamoxifen 20<br />

mg/day while postmenopausal women with positive receptors were assigned to<br />

receive 5 years <strong>of</strong> sequential aromatase inhibitor (AI) and tamoxifen.<br />

Follow-Up: After completion <strong>of</strong> chemotherapy and radiotherapy, patients were<br />

planned for follow up visits every 3 months during the first year, every 4 months<br />

in the second year, and every 6 months until the end <strong>of</strong> the fifth year. Patients were<br />

subjected to careful history-taking and physical examination. A mammogram<br />

and echocardiography were performed yearly whereas, chest X-ray, abdominal<br />

ultrasound or bone scan were requested if symptoms or laboratory abnormalities<br />

were present.<br />

Outcome Measures: The primary end point <strong>of</strong> this study was relapse-free<br />

survival (RFS) which was defined as the time from surgery to documented<br />

recurrence. Local breast recurrence, nodal recurrence, and metastatic disease<br />

were considered as a recurrence. Patients who had a contralateral breast cancer<br />

or a second malignancy were not considered to have a relapse. Secondary<br />

outcome measures included overall survival (OS) which was defined as the time<br />

from surgery to documented death due to any cause. Treatment-related toxicity<br />

is another secondary end point.<br />

AC/T versus FAC analysis: The treatment outcome <strong>of</strong> dose-dense AC/T was<br />

eventually compared with the available treatment results <strong>of</strong> the conventionally<br />

treated high-risk node-positive patients using adjuvant 6 cycles <strong>of</strong> FAC<br />

combination chemotherapy (5-flurouracil 600mg/m2 , doxorubicin 60 mg/m2 and<br />

cyclophosphamide 600 mg/m2 were administered IV on day 1) scheduled every<br />

3 weeks. Patients received FAC chemotherapy enrolled in the analysis had the<br />

same inclusion criteria and were treated during a defined period <strong>of</strong> time with a<br />

comparable follow-up period to assess the same outcome measures. The relevant<br />

data including the clinico-pathological characteristics, outcome parameters and<br />

treatment-related toxicity were obtained from the medical records.<br />

Statistical analysis: Data were statistically described in terms <strong>of</strong> range, mean ±<br />

standard deviation (± SD), frequencies (number <strong>of</strong> cases) and percentages when<br />

appropriate. Survival analysis was done for the different outcome measures<br />

using Kaplan Maier statistics calculating the mean and median survival time for<br />

each group with their 95% CI and the corresponding survival graphs. Factors<br />

examined as a stepwise adjusted Kaplan Meier were age at presentation (<br />

35, 35 - 49 , 50 years), T status (T1, T2, T3, T4), N status (N1, N2, N3),<br />

menopausal status (postmenopausal or premenopausal), ER status (negative or<br />

positive), PR status (negative or positive), and HER2/neu status (negative or<br />

amplified). A plot <strong>of</strong> RFS and OS Vs the mentioned factors were calculated and<br />

compared between the two groups. A probability value (p value) less than 0.05<br />

was considered statistically significant. All statistical calculations were done<br />

using computer programs Micros<strong>of</strong>t Excel 2007 (Micros<strong>of</strong>t Corporation, NY,<br />

USA) and SPSS (Statistical Package for the Social Science; SPSS Inc., Chicago,<br />

IL, USA) version 17 for Micros<strong>of</strong>t Windows.<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 35<br />

Results<br />

A total <strong>of</strong> 61 patients were recruited to receive dose dense AC-T and were<br />

compared with 43 patients received standard FAC chemotherapy with respect<br />

to the clinico-epidemiological features, disease-related events and treatmentrelated<br />

events. The follow-up period ranged from 12 months to 48 months with a


original article <<br />

median follow up <strong>of</strong> 37 months. Baseline characteristics were balanced between<br />

both treatment arms with no statistically significant differences. (Table 1)<br />

Table 1: Baseline patient and tumor characteristics<br />

FAC<br />

(no.=43)<br />

AC-T<br />

(no.=61)<br />

P<br />

value<br />

No. % No. %<br />

Age (years)<br />

35 3 7 8 13.1<br />

36-50 21 48.8 25 41<br />

50+ 19 44.2 28 45.9 0.53<br />

Median (range)<br />

Menopausal<br />

status<br />

47.5 (27 - 65) 47 (24 -63) 0.6<br />

Pre menop. 20 46.5 38 62.3<br />

Post menop.<br />

Grade<br />

23 53.5 23 37.7 0.81<br />

G1 2 4.65 2 3.3<br />

G2 39 90.70 54 91.5<br />

G3<br />

Type <strong>of</strong> surgery<br />

2 4.65 5 8.2 0.2<br />

MRM 32 74.5 54 88.5<br />

BCS<br />

T-stage<br />

11 25.5 7 11.5 0.16<br />

T1 5 11.6 8 13.1<br />

T2 26 60.4 39 63.9<br />

T3 5 11.6 10 16.4<br />

T4<br />

N-stage<br />

7 16.4 4 6.6 0.41<br />

N1 3 7 6 9.8<br />

N2 23 53.5 33 54.1<br />

N3<br />

Receptor status<br />

17 39.5 22 36.1 0.85<br />

ER +ve 35 81.4 44 72.1<br />

ER -ve 8 18.6 17 27.9 0.19<br />

PR +ve 33 76.7 41 67.2<br />

PR -ve<br />

HER2/neu<br />

10 23.3 20 32.8 0.22<br />

Negative 38 88.4 51 83.6<br />

Positive 5 11.6 10 16.4 0.76<br />

Fifty patients out <strong>of</strong> the 61 patients (81.9%) in the dose dense arm completed the<br />

8 cycles <strong>of</strong> chemotherapy within 110 days (planned time between the first and the<br />

last cycles). The delay in the majority <strong>of</strong> patients was mainly due to treatmentrelated<br />

toxicity (neuropathy, cytopenia and febrile neutropenia) and rarely<br />

patient non-compliance. Dose reduction due to grade 3-4 toxicity was employed<br />

in 8 patients (13.1%). On the other hand, 39 out <strong>of</strong> the 43 patients (90.6%) in the<br />

FAC arm completed the 6 cycles within 110 days and most <strong>of</strong> the treatment delay<br />

was due to febrile neutropenia. Dose reduction due to hematological toxicity was<br />

required only in one patient (2.8%). The mean chemotherapy duration was 123<br />

± 5.9 days in the dose dense AC/T arm compared to 117 ± 6.3 days in the FAC<br />

arm and (P= 0.05).<br />

The 3-year adjusted RFS rates for AC-T and FAC were 76 % and 54.6 %<br />

respectively (P=0.042) (Fig.1). The mean disease-free interval was 40.6 ± 7.2<br />

months (95% CI, 37.7 - 43.6) for dose-dense AC/T arm Vs 36.9 ± 6.9 months<br />

(95% CI, 32.3 - 41.5) for FAC arm, (P=0.040). Regarding the pattern <strong>of</strong> relapse,<br />

systemic relapse (metastatic disease) was the predominant pattern in both arms<br />

(87.8% for dose-dense AC/T arm and 78.6% for FAC arm).<br />

Fig. 1: Kaplan-Maier survival curve <strong>of</strong> relapse-free survival in both<br />

treatment arms<br />

The subgroup analysis revealed that dose-dense chemotherapy had a statistically<br />

significant positive effect on 3-year RFS in premenopausal patients (77.5%<br />

for AC/T arm Vs 54% for FAC arm, P=0.045), patients with 10 or more (N3)<br />

positive axillary lymph nodes (72% for AC/T arm Vs 26% for FAC arm, P=0.02)<br />

and positive ER status (83% for AC/T arm Vs 50% for FAC arm, P=0.017).<br />

HER2/neu status did not significantly affect the 3-year RFS in patients received<br />

dose-dense AC/T though there was a trend towards better outcome compared<br />

with FAC arm in HER2/neu positive patients (77% for AC/T arm Vs 57% for<br />

FAC arm, P=0.051),<br />

There was no statistically significant difference in the 3-year OS between the two<br />

regimens or subgroups (Fig. 2). Mean OS was 46.1 ± 5.6 months for FAC arm<br />

compared to 43 ± 6.0 months in the dose dense arm (P= 0.7). Two patients in the<br />

FAC arm and 3 patients in the AC/T arm died due to extensive metastatic disease<br />

(mainly pulmonary and liver metastases).<br />

36 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


Fig. 2: Kaplan-Maier survival curve <strong>of</strong> overall survival in both treatment<br />

arms<br />

Hematologic toxicity was more pronounced in the dose dense arm. The difference<br />

was highly significant (P=0.01) with respect to anemia, granulocytopenia and<br />

thrombocytopenia. In the AC/T arm, the incidence <strong>of</strong> hematological toxicity<br />

was more evident during treatment with paclitaxel rather than AC. All patients<br />

had full recovery <strong>of</strong> their peripheral-blood counts at the end <strong>of</strong> treatment. No<br />

toxicity-related mortality was observed in both arms. Non hematologic toxicity<br />

occurred significantly more <strong>of</strong>ten in the dose dense arm. For example, peripheral<br />

sensory neuropathy grade 3-4 was more evident in patients received AC/T 36%<br />

Vs 2.3% in FAC arm (P=0.002) (Table 2). After a median follow-up period <strong>of</strong><br />

more than 3 years, only 2% <strong>of</strong> the patients received dose-dense chemotherapy<br />

had persistent grade 1 or 2 neurotoxicity compared with none in the FAC arm.<br />

In both arms, none <strong>of</strong> the patients developed clinically evident congestive heart<br />

failure or significant reduction in the ejection fraction. Furthermore, there<br />

were no recorded cases <strong>of</strong> secondary malignancy, leukemia or myelodysplastic<br />

syndrome (MDS)<br />

Table 2: Treatment-related toxicity (G III/IV) in both arms<br />

Toxicity FAC arm AC-T arm<br />

No. % No. %<br />

Nausea 25 58 50 81<br />

Vomiting 17 39.5 36 59<br />

Granulocytopenia 9 20.9 35 57<br />

Thrombocytopenia 4 9.3 3 5<br />

Anemia 6 13.9 30 49<br />

Neuropathy 1 2.3 22 36<br />

Febrile neutropenia 1 2.3 3 5<br />

Cardiotoxicity 0 0 0 0<br />

Fatigue 15 34.8 15 24.5<br />

Bony pains 2 4.6 3 4.9<br />

Discussion<br />

The outcomes <strong>of</strong> women with early breast cancer have improved incrementally<br />

through trials that have evaluated new chemotherapy drugs and different doses<br />

and schedules <strong>of</strong> drugs postoperatively. Although much progress has been made,<br />

women still develop recurrences (15). Dose-dense chemotherapy has become<br />

one <strong>of</strong> the possible standards <strong>of</strong> adjuvant chemotherapy (16). The hypothesis that<br />

such a strategy is effective was based, in part, on theories developed by Skipper<br />

and by Norton and Simon (17-19). In their experimental models, a given dose <strong>of</strong><br />

drug always kills a certain fraction, rather than a certain number, <strong>of</strong> exponentially<br />

growing cancer cells. However, breast cancer cells proliferate by nonexponential<br />

gompertzian kinetics, and the rate <strong>of</strong> cancer cell proliferation between treatment<br />

cycles is more rapid than that used in exponential models (19).<br />

The current study was designed to assess the treatment outcome <strong>of</strong> adjuvant<br />

dose-dense AC/T in high-risk breast cancer patients and to identify the possible<br />

subgroup <strong>of</strong> patients that had a significant improvement <strong>of</strong> their survival<br />

parameters. Due to the growing evidence from recent clinical trials that<br />

showed improvement <strong>of</strong> treatment outcome in favor <strong>of</strong> dose-dense compared<br />

to conventionally scheduled chemotherapy, prospective data <strong>of</strong> AC/T were<br />

compared with the available data <strong>of</strong> the classic FAC regimen used to be the<br />

standard adjuvant treatment for high-risk patients. Baseline characteristics were<br />

balanced between both treatment arms with no statistically significant differences.<br />

The results <strong>of</strong> the current study revealed that dose-dense AC/T was superior to<br />

conventional FAC in terms <strong>of</strong> RFS. The benefit was evident in premenopausal<br />

patients, patients with 10 or more (N3) positive axillary lymph nodes and<br />

positive ER status, whereas the benefit was not affected by the HER2/neu status.<br />

There was no statistically significant difference in the 3-year OS between the two<br />

regimens. Several clinical trials have confirmed the importance <strong>of</strong> dose density<br />

and dose intensity in the adjuvant treatment <strong>of</strong> early breast cancer, especially in<br />

women with positive nodal status.<br />

The Cancer and Leukemia Group B (CALGB) trial C9741 compared 4 cycles <strong>of</strong><br />

AC followed by 4 cycles <strong>of</strong> paclitaxel in dose-dense intervals with conventional<br />

3-week intervals in 1,973 node-positive patients and found that the dosedense<br />

regimen produced significantly better DFS (HR=0.74; P=.010) and OS<br />

(HR=0.69; p=.013). Severe neutropenia was observed less frequently in patients<br />

receiving G-CSF support with the dose-dense chemotherapy schedule, and the<br />

regimen was otherwise well tolerated (12). This finding was recently challenged<br />

by an extensive retrospective analysis <strong>of</strong> this and two other CALGB trials, in<br />

which patients who were estrogen or progesterone receptor positive and received<br />

tamoxifen after chemotherapy experienced a markedly smaller effect <strong>of</strong> dosedense<br />

delivery <strong>of</strong> chemotherapy, with a minimal reduction in the relative risk for<br />

recurrence and no reduction in the risk for death (20).<br />

The German Arbeitsgemeinschaft Gastrointestinal Onkologie (AGO) trial<br />

examined 1,284 patients with more than four positive lymph nodes using<br />

treatment with a dose-dense, dose-intense regimen <strong>of</strong> epirubicin, paclitaxel, and<br />

cyclophosphamide (E-T-C) administered sequentially on a 2-week schedule with<br />

GCSF support. These very high risk patients achieved 2- year RFS rates <strong>of</strong> 85%,<br />

versus 82% for conventionally dosed EC + T (p = .046, two-tailed test). Survival<br />

benefits were observed irrespective <strong>of</strong> hormone receptor status or HER-2<br />

expression, but were particularly impressive in women with 10 or more positive<br />

lymph nodes. That trial used epirubicin doses <strong>of</strong> 150 mg/m2 every 2 weeks for a<br />

cumulative epirubicin dose <strong>of</strong> 450 mg/m2 in the dose-dense arm (21).<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 37


original article <<br />

Many studies have suggested that the benefit <strong>of</strong> taxane-based or dose-dense<br />

chemotherapy may be restricted to patients with HER2-positive or steroid<br />

hormone receptor– negative disease (22, 23). The current study failed to<br />

demonstrate such benefit as the dose-dense regimen had a positive effect in<br />

patients with estrogen receptor–positive breast cancer with no effect <strong>of</strong> HER2/<br />

neu status. The possible explanation could be the higher frequency <strong>of</strong> N3 disease<br />

(36%) among patients received AC/T, which may have minimized the effect <strong>of</strong><br />

biological predictors <strong>of</strong> response (HER2/neu status) and maximized the effect <strong>of</strong><br />

clinico-pathological factors (nodal status).<br />

However, not all adjuvant dose-dense trial data were viewed as positive. The<br />

GONO-MIG (Gruppo Oncologico Nord Ovest- Mammella InterGruppo)<br />

trial compared FU, epirubicin, and cyclophosphamide administered every 3<br />

weeks versus the same regimen given every 2 weeks with granulocyte colonystimulating<br />

factor support. As in the CALGB study, the chemotherapy dose<br />

per cycle and total number <strong>of</strong> cycles were held constant, and only the interval<br />

between cycles varied. The difference between these two arms did not reach<br />

statistical significance for either recurrence or death (24). These negative results<br />

may be explained by the study design. The GONO-MIG trial applied a suboptimal<br />

version <strong>of</strong> the FEC regimen (ie, FU at 600 mg/m2, epirubicin at 60 mg/m2, and<br />

cyclophosphamide at 600 mg/m2_six cycles). The total dose <strong>of</strong> epirubicin in<br />

both arms was 360 mg/m2, which corresponded to only 50% <strong>of</strong> the total<br />

dose in the highly effective Canadian CEF regimen (ie, cyclophosphamide 75<br />

mg/m2 orally days 1 through 14, epirubicin 60 mg/m2 days 1 and 8, and FU<br />

500mg/m2 intravenously day 1and 8 for six cycles) or to only 60% <strong>of</strong> the total<br />

dose in the French FE100C regimen (FU500mg/m2, epirubicin 100mg/m2, and<br />

cyclophosphamide 500 mg/m2 for six cycles) (25, 26).<br />

Hematologic and non hematologic toxicities were more pronounced in the<br />

dose-dense AC/T. However, all patients had full recovery <strong>of</strong> their peripheralblood<br />

counts at the end <strong>of</strong> treatment and only 2% <strong>of</strong> the patients had persistent<br />

grade 1 or 2 neurotoxicity. At a median follow up <strong>of</strong> 37 months, there were<br />

no recorded cases <strong>of</strong> secondary malignancy, leukemia or MDS and this finding<br />

does not correspond to published data <strong>of</strong> anthracycline and cyclophosphamide<br />

adjuvant regimens. In the randomized, phase III trial, conducted by Moebus et al,<br />

1,284 patients with four or more involved axillary lymph nodes were randomly<br />

assigned to receive intense dose-dense (IDD) sequential epirubicin, paclitaxel,<br />

and cyclophosphamide (IDD-ETC) every 2 weeks or conventionally scheduled<br />

epirubicin/cyclophosphamide followed by paclitaxel every three weeks. Four<br />

occurrences (0.6% <strong>of</strong> patients) <strong>of</strong> secondary leukemia/MDS were reported with<br />

the 5-year follow-up data in the IDD-ETC arm only (16).<br />

With 10 years <strong>of</strong> follow-up, 5 occurrences (1.4% <strong>of</strong> patients) <strong>of</strong> secondary<br />

leukemia/MDS were reported for patients receiving the Canadian CEF<br />

regimen (25). Praga et al, reviewed 19 adjuvant trials with epirubicin and<br />

cyclophosphamide in 2005. Depending on the total dose <strong>of</strong> both epirubicin and<br />

cyclophosphamide, patients had a 8-year cumulative probability <strong>of</strong> secondary<br />

leukemia/MDS ranging between 0.37% and 4.97% (27). The absence <strong>of</strong><br />

secondary leukemia/MDS in the current study may be explained by the relatively<br />

short follow- up interval and low total anthracycline dose (240 mg/m2 in the<br />

dose dense arm and 360 mg/m2 in the conventional arm).<br />

In conclusion, dose dense AC/T is a highly effective, feasible, and safe regimen<br />

with manageable toxicity for adjuvant treatment <strong>of</strong> node-positive high-risk<br />

breast cancer patients. Future research should focus on predictive factors for<br />

different chemotherapeutic regimens and on combining targeted therapies with<br />

dose-dense regimens to continue the incremental advance in the outcomes <strong>of</strong><br />

women with early breast cancer.<br />

Conclusion<br />

Dose-dense AC/T significantly improved the relapse-free survival in patients<br />

with high-risk primary breast cancer and was less well tolerated compared with<br />

the conventionally scheduled FAC. The benefit was evident in premenopausal<br />

patients, extensive axillary nodal metastasis and positive ER status.<br />

References<br />

1. Levine MN, Whelan T: Adjuvant chemotherapy for breast cancer: 30<br />

years later. N Engl J Med 2006; 355:1920-1922.<br />

2. Levine MN, Eisen A: Anthracycline adjuvant chemotherapy: How much<br />

is enough? J Clin Oncol 2001; 19:599-601.<br />

3. Dang C, Hudis C: Adjuvant taxanes in the treatment <strong>of</strong> breast cancer: No<br />

longer at the tip <strong>of</strong> the iceberg. Clin Breast Cancer 2006; 7:51-58.<br />

4. 4. Hryniuk W, Levine MN: Analysis <strong>of</strong> dose intensity for adjuvant<br />

chemotherapy trials in stage 2 breast cancer. J Clin Oncol 1986; 4:1162-<br />

1170.<br />

5. Norton L: A Gompertzian model <strong>of</strong> human breast cancer growth. Cancer<br />

Res 1988; 48: 7067-7071.<br />

6. Skipper HE: Kinetics <strong>of</strong> mammary tumor cell growth and implications for<br />

therapy. Cancer 1971; 28: 1479-1499.<br />

7. Levine MN, Bramwell VH, Pritchard KI, et al: Randomized trial <strong>of</strong><br />

intensive cyclophosphamide, epirubicin, and fluorouracil chemotherapy<br />

compared with cyclophosphamide, methotrexate, and fluorouracil in<br />

premenopausal women with node-positive breast cancer: National Cancer<br />

Institute <strong>of</strong> Canada Clinical Trials Group. J Clin Oncol 1998; 16:2651-<br />

2658.<br />

8. Therasse P, Mauriac L, Welnicka-Jaskiewicz M, et al: Final results <strong>of</strong><br />

a randomized phase III trial comparing cyclophosphamide, epirubicin, and<br />

fluorouracil with a dose-intensified epirubicin and cyclophosphamide _<br />

filgrastim as neoadjuvant treatment in locally advanced breast cancer: An<br />

EORTC-NCIC-SAKK multicenter study. J Clin Oncol 2003;21:843-850.<br />

9. Roche´ H, Fumoleau P, Spielmann M, et al: Sequential adjuvant epirubicinbased<br />

and docetaxel chemotherapy for node-positive breast cancer<br />

patients: The FNCLCC PACS 01 Trial. J Clin Oncol 2006; 24:5664-5671.<br />

10. Francis P, Crown J, Di Leo A, et al: Adjuvant chemotherapy with<br />

sequential or concurrent anthracycline and docetaxel: Breast International<br />

Group 02-98 randomized trial. J Natl Cancer Inst 2008; 100:121-133.<br />

11. Mamounas EP, Bryant J, Lembersky BC, et al: Paclitaxel after<br />

doxorubicin plus cyclophosphamide as adjuvant chemotherapy for nodepositive<br />

breast cancer: Results from NSABP-B 28. J Clin Oncol 2005;<br />

23:3686-3696.<br />

12. Citron ML, Berry DA, Cirrincione C, et al: Randomized trial <strong>of</strong> dosedense<br />

versus conventionally scheduled and sequential versus concurrent<br />

combination chemotherapy as postoperative adjuvant treatment <strong>of</strong> nodepositive<br />

primary breast cancer: First report <strong>of</strong> intergroup trial C9741/Cancer<br />

and Leukemia Group B Trial 9741. J Clin Oncol 2003; 21:1431-1439.<br />

13. Hudis C, Citron M, Berry D, et al: Five year follow-up <strong>of</strong> INT C9741:<br />

Dose-dense chemotherapy is safe and effective. Breast Cancer Res Treat<br />

2005; 94: S20, (suppl; abstr 41).<br />

38 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


14. National Cancer Institute: Common Toxicity Criteria Manual, version<br />

2.0, June 1, 1999. http:/ ctep.cancer.gov/reporti / ctc. Html.<br />

15. Burnell M, Levine MN, Chapman JA, et al: Cyclophosphamide,<br />

epirubicin, and fluorouracil versus dose-dense epirubicin and<br />

cyclophosphamide followed by paclitaxel versus dDoxorubicin and<br />

cyclophosphamide followed by paclitaxel in node-positive or high-risk<br />

node-negative bBreast cancer. J Clin Oncol 2009; 28:77-82.<br />

16. Moebus V, Jackisch C, Lueck HJ, et al: Intense dose-dense sequential<br />

chemotherapy with epirubicin, Paclitaxel, and cyclophosphamide<br />

compared with conventionally scheduled chemotherapy in high-risk<br />

primary breast cancer: Mature results <strong>of</strong> an AGO phase III study. J Clin<br />

Oncol 2010; 28:2874-2880.<br />

17. Skipper HE: Laboratory models: some historical perspective. Cancer<br />

Treat Rep 1986; 70 : 3 – 7.<br />

18. Norton L, Simon R, Brerenton JD, et al: Predicting the course <strong>of</strong><br />

Gompertzian growth. Nature 1976; 264 : 542 – 5.<br />

19. Norton L: Theoretical concepts and the emerging role <strong>of</strong> taxanes in<br />

adjuvant therapy. Oncologist 2001; 3 : 30 – 5 (suppl).<br />

20. Berry DA, Cirrincione C, Henderson IC et al: Effects <strong>of</strong> improvements<br />

in chemotherapy on disease-free and overall survival <strong>of</strong> estrogen-receptor<br />

negative, node-positive breast cancer: 20-year experience <strong>of</strong> the CALGB<br />

& U.S. Breast Intergroup. Breast Cancer Res Treat 2004; 88:S17.<br />

21. Moebus V, Untch M, Du Bois A et al : Dose-dense sequential<br />

chemotherapy with epirubicin (E), paclitaxel (T) and cyclophosphamide<br />

(C) (ETC) is superior to conventional dosed chemotherapy in high-risk<br />

breast cancer patients (≥4 + LN). First results <strong>of</strong> an AGO-trial. Proc Am<br />

Soc Clin Oncol 2004; Late-Breaking Abstracts Book: 14.<br />

22. Hayes DF, Thor AD, Dressler LG, et al: HER2 and response to paclitaxel<br />

in node-positive breast cancer. N Engl J Med 2007; 357:1496-1506.<br />

23. Berry DA, Cirrincione C, Henderson IC, et al: Estrogen-receptor status<br />

and outcomes <strong>of</strong> modern chemotherapy for patients with node-positive<br />

breast cancer. JAMA 2006; 295:1658-1667.<br />

24. Venturini M, Del Mastro L, Aitini E, et al: Dose-dense adjuvant<br />

chemotherapy in early breast cancer patients: Results from a randomized<br />

trial.J Natl Cancer Inst 2005; 97:1724-1733.<br />

25. Levine MN, Pritchard KI, Bramwell VHC, et al: Randomized<br />

trial comparing cyclophosphamide, epirubicin, and fluorouracil with<br />

cyclophosphamide, methotrexate, and fluorouracil in premenopausal<br />

women with node-positive breast cancer: Update <strong>of</strong> National Cancer<br />

Institute <strong>of</strong> Canada Clinical Trials Group Trial MA5. J Clin Oncol 2005;<br />

23:5166-5170.<br />

26. Bonneterre J, Roche H, Kerbrat P et al: Epirubicin increases longterm<br />

survival in adjuvant chemotherapy <strong>of</strong> patients with poor-prognosis,<br />

nodepositive, early breast cancer: 10-year follow-up results <strong>of</strong> the<br />

French Adjuvant Study Group 05 randomized trial J Clin Oncol 2005;<br />

23(12):2686-93<br />

27. Praga C, Bergh J, Bliss J, et al: Risk <strong>of</strong> acute myeloid leukemia and<br />

myelodysplastic syndrome in trials <strong>of</strong> adjuvant epirubicin for early breast<br />

cancer: Correlation with doses <strong>of</strong> epirubicin and cyclophosphamide. J Clin<br />

Oncol 2005; 23:4179-4191.<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 39


original article <<br />

Genomics views: Xenobiotic metabolizing enzymes and cancer risks<br />

Kamel Rouissi, MD* 1 , Bechr Hamrita, MD* 1 , Slah Ouerhani, MD 2 , Amel Benammar Elgaaied, MD 1<br />

(1) Laboratory <strong>of</strong> Genetic, Immunology and Human Pathology, Faculty <strong>of</strong> Sciences <strong>of</strong> Tunis, University <strong>of</strong> El Manar I. 2092,<br />

Tunis, Tunisia<br />

(2) Laboratory <strong>of</strong> Molecular and Cellular Hematology, Institut Pasteur de Tunis, Tunisia<br />

* Both authors contributed equally to the study.<br />

Corresponding Author: Correspondence to: Dr. Bechr Hamrita, MD<br />

Laboratory <strong>of</strong> Genetics, Immunology and Human Pathology, Faculty <strong>of</strong> sciences <strong>of</strong> Tunis<br />

University <strong>of</strong> El Manar I, 2092, Tunis, Tunisia.<br />

E-mai: bechrhamrita@yahoo.fr<br />

Keywords: Cytochrome P450, N-acetyltransferase, Glutathione S-transferase, Cancer, Polymorphisms, Xenobiotic enzymes.<br />

ISSN: 2070-254X<br />

Abstract<br />

Epidemiological studies have estimated that approximately 80% <strong>of</strong> all cancers<br />

are related to environmental factors. Individual cancer susceptibility can be<br />

the result <strong>of</strong> several host factors, including differences in metabolism, DNA<br />

repair, altered expression <strong>of</strong> tumor suppressor genes and proto-oncogenes, and<br />

nutritional status. In fact, xenobiotic metabolism is the principal mechanism<br />

for maintaining homeostasis during the body’s exposure to xenobiotics. Most<br />

xenobiotics that enter in the body are subjected to metabolism that functions<br />

primarily to facilitate their elimination. Metabolism <strong>of</strong> certain xenobiotics<br />

can also result in the production <strong>of</strong> electrophilic derivatives that can cause<br />

cell toxicity and transformation. The balance <strong>of</strong> xenobiotic absorption and<br />

elimination rates in metabolism can be important in the prevention <strong>of</strong> DNA<br />

damage by chemical carcinogens. Thus the ability to metabolize and eliminate<br />

xenobiotics can be considered one <strong>of</strong> the body’s first protective mechanisms.<br />

However, there are marked species differences in the way mammals respond to<br />

xenobiotics, which are due in large part to molecular differences in xenobioticmetabolizing<br />

enzymes and has been related to the enzymatic polymorphisms<br />

involved in activation and detoxification <strong>of</strong> chemical carcinogens and<br />

that can impact drug therapy and cancer susceptibility. This paper focus<br />

the member <strong>of</strong> the cytochrome P450 family <strong>of</strong> enzymes (Cyp2D6), glutathione<br />

S-transferases (GSTT1 and GSTM1 ) and N-acetyltransferases (NAT1 and<br />

NAT2) on genetic polymorphisms involved in the metabolism <strong>of</strong> endocrine<br />

disruptors potentially related to cancer development.<br />

Introduction<br />

Xenobiotics are natural or artificial chemical substances that are alien to the body,<br />

such as drugs, industrial products, pollutants, alkaloids, and toxins produced<br />

by fungi, plants, and animals, many <strong>of</strong> them acting as endocrine disruptors.<br />

In their natural or bio-transformed state, xenobiotics can affect DNA integrity,<br />

leading to cancer if exposure persists (1). Accumulated DNA damage, added<br />

to spontaneous replication errors not corrected by the repair system, can cause<br />

irreversible mutations which in turn can lead to the development <strong>of</strong> tumors and/<br />

or progression <strong>of</strong> cancer. Epidemiological studies show that 80 % <strong>of</strong> all cancers<br />

are related to environmental factors like smoking and occupational and dietary<br />

exposures (2). Thus, the individual capacity to bio-transform toxic into non-toxic<br />

xenobiotics can be considered the first line <strong>of</strong> defense in the process characterized<br />

by successive stages <strong>of</strong> transformation <strong>of</strong> potentially toxic chemical substances<br />

as a pathway towards their subsequent elimination. The enzymes involved are<br />

frequently the ones that determine the intensity and duration <strong>of</strong> drug action and<br />

other xenobiotics, hence their importance in chemical and carcinogenic toxicity.<br />

Bio-transformation <strong>of</strong> xenobiotics involves the modification <strong>of</strong> their physical<br />

properties, generally from lipophilic to hydrophilic, facilitating their excretion.<br />

Otherwise, many lipophilic xenobiotics would be excreted so slowly that they<br />

would eventually accumulate, destroying the organism by making it biologically<br />

nonviable (1). Bio-transformation involves two stages: phase I, mainly<br />

involving enzymatic activity <strong>of</strong> the cytochrome P450 (CYP) family; and phase<br />

II, catalyzed by conjugation enzymes like glutathione S-transferase (GST), and<br />

N-acetyltransferase (NAT).<br />

Most carcinogenic chemical products are not toxic and require metabolic<br />

activation before interacting with cellular macro-molecules. Phase I enzymes<br />

promote the activation <strong>of</strong> drugs and pro-carcinogens for the genotoxic<br />

electrophilic intermediaries. Meanwhile, phase II enzymes generally act<br />

as inactivating enzymes, that is, they catalyze the binding <strong>of</strong> intermediary<br />

metabolites to c<strong>of</strong>actors, transforming them into more hydrophilic products,<br />

thus facilitating their elimination (3). Therefore, the coordinated expression<br />

and regulation <strong>of</strong> xenobiotic metabolizing enzymes (XMEs) in both phase I<br />

and phase II and their metabolic equilibrium in the cells <strong>of</strong> target organs can be<br />

important factors in determining susceptibility to cancer as related to exposure<br />

to carcinogens (4). Mutations in these genes can produce partially defective<br />

enzymes or ones with altered specificities to the substrates, and thus there may or<br />

may not be the production <strong>of</strong> functional proteins or even enzymes with different<br />

levels <strong>of</strong> activities. The combination <strong>of</strong> the alleles from these genes can cause an<br />

increase or decrease in the susceptibility to certain toxic agents or environmental<br />

carcinogens.<br />

There are marked inter-individual and inter-ethnic differences in the capacity<br />

to metabolize drugs and other xenobiotics. This variation is due to the<br />

polymorphisms in the corresponding genes and to physiological, pathological,<br />

and environmental factors (5). Inter-individual variability in xenobiotic<br />

metabolism has been associated with greater or lesser susceptibility to toxicity or<br />

cancer risk in response to the same exposure to a given environmental pollutant.<br />

Thus, individuals incapable <strong>of</strong> adequately detoxifying a metabolic carcinogen<br />

40 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


or toxic agent due to reduced enzymatic activity would undergo more DNA<br />

and cell damage with the formation <strong>of</strong> adducts, or chemical elements bound to<br />

the DNA and protein macromolecules, genomic instability, and consequently<br />

would have a greater risk <strong>of</strong> developing toxicity or cancer (3). Increasing<br />

attention has been focused on our knowledge <strong>of</strong> variations in susceptibility to<br />

diseases within a population and the identification <strong>of</strong> risk factors so as to orient<br />

preventive policies. The literature has shown that variability in the expression<br />

<strong>of</strong> genes for XMEs (xenobiotic metabolizing enzymes) suggests an influence<br />

on the biological response to carcinogens. Despite displaying weak indication<br />

<strong>of</strong> risk at the individual level due to association with various other factors,<br />

gene polymorphisms (principally those influencing the metabolic activation or<br />

detoxification <strong>of</strong> carcinogenic chemical products) can be important factors for<br />

susceptibility at the population level (6). Molecular epidemiology has made great<br />

progress in detecting many human gene polymorphisms in XMEs, and some<br />

have shown to be correlated to increased risk <strong>of</strong> cancer. This paper focuses on<br />

just a few genetic polymorphisms that have been investigated more extensively<br />

due to their association with cancer, such as genes encoding cytochrome P450<br />

(CYP2D6), glutathione S-transferase (GSTM1 and GSTT1) families and the<br />

N-acetyltransferases (NAT1 and NAT2).<br />

Cytochrome P450 (CYPs)<br />

The human cytochrome P450 superfamily comprises at least ten known and<br />

characterized families and numerous sub-families (7). During bio-transformation,<br />

cytochrome P450 mediates the phase I reactions in which xenobiotics are<br />

detoxified or activated to reactive intermediate substances. The highest<br />

concentration <strong>of</strong> these enzymes has been observed in the liver endoplasmic<br />

reticulum, but they are present in all tissues in a tissue-specific manner. In the<br />

liver, they determine the intensity and duration <strong>of</strong> drug action and promote the<br />

detoxification <strong>of</strong> xenobiotics. They also catalyze the activation <strong>of</strong> xenobiotics to<br />

toxic and/or carcinogenic metabolites. The contribution <strong>of</strong> each P450 enzyme to<br />

the activation <strong>of</strong> carcinogens has been extensively evaluated, and this research<br />

has shown that most environmental carcinogens are activated principally by a<br />

limited number <strong>of</strong> them, including the following: CYP1A1, CYP1A2, CYP2E1,<br />

CYP2C19, CYP2D6, and CYP3A (8). Many are polymorphic, displaying<br />

different metabolic activities, reflected in adverse toxic effects, including<br />

carcinogenesis induced by endogenous chemical substances (9).<br />

CYP2D6<br />

The CYP2D6 gene belongs to the CYP2 family and was mapped in human<br />

chromosome 22, band 22q13.1 (10). Enzyme CYP2D6 (debrisoquine-4hydroxylase)<br />

metabolizes debrisoquine and many other drugs, like antidepressants,<br />

neuroleptics, many anti-arrhythmics, and lipophilic b-blockers (9).<br />

In addition to these substrates, CYP2D6 also acts on the carcinogen nitrosamine<br />

NNK (4-methylnitrosamino-1(3-pyridyl)-1-butanone), a component <strong>of</strong> cigarette<br />

smoke (11). The absence <strong>of</strong> debrisoquine-4-hydroxylase activity can have<br />

serious clinical consequences and even lead to death, since usual doses can<br />

cause high plasma levels <strong>of</strong> the drug, leading to side effects (12). Debrisoquine<br />

is a drug used for treating hypertension, and a wide variation has been observed<br />

in the hypotensive response. A clinical consequence <strong>of</strong> slow metabolism is the<br />

great sensitivity to the anti-hypertensive effects <strong>of</strong> debrisoquine (13). Although,<br />

extensive metabolizers display less risk <strong>of</strong> the effects <strong>of</strong> overdoses from<br />

debrisoquine and related drugs, they show an increased risk <strong>of</strong> developing cancer<br />

<strong>of</strong> the liver, gastro-intestinal tract, and lung as compared to slow metabolizers (14).<br />

Most individuals (80-90%) have at least one wild allele (CYP2D6*1) for the<br />

CYP2D6 gene and are classified functionally as extensive metabolizers. There are<br />

two other groups <strong>of</strong> individuals: one with intermediate metabolic activity, known<br />

as intermediate metabolizers, and the other known as ultra-rapid metabolizers.<br />

The first phenotype is attributed to a mutation in the wild allele (CYP2D6*1) and<br />

the second to an amplification <strong>of</strong> either the wild allele or an active mutant allele.<br />

Finally, there is a small group (5-10% <strong>of</strong> Caucasians, 2% <strong>of</strong> African Americans,<br />

and 1% <strong>of</strong> Orientals) who are poor metabolizers and identified by loss <strong>of</strong> gene<br />

function and absence <strong>of</strong> protein (15).<br />

An updated review <strong>of</strong> this complex polymorphism is provided by Sachse et al,<br />

(16). According to these authors, different alleles for the CYP2D6 gene consist<br />

mainly <strong>of</strong> point mutations, conversions, gene duplications, and complete gene<br />

deletion. Some 15 alleles have been recorded and associated with low activity<br />

(CYP2D6*2, *9, *10) and with its absence (CYP2D6*3, *4, *5, *6, *7, *8, *11,<br />

*12, *13, *14, *15, *16). The combination <strong>of</strong> all these alleles provides a wide<br />

range <strong>of</strong> possible phenotypes in relation to CYP2D6 activity. Given the nature<br />

<strong>of</strong> the substances metabolized by these enzymes, this polymorphism is used<br />

principally to identify poor metabolizers with anomalous responses to given<br />

drugs. In ultra-rapid metabolizers, the usual doses <strong>of</strong> given drugs fail to produce<br />

the desired pharmacological effect. Determination <strong>of</strong> CYP2D6 expression<br />

serves to detect therapeutic problems due to metabolism and can contribute<br />

to individualization <strong>of</strong> the dose regimen, reaching optimum drug therapeutic<br />

levels and reducing both cost and possible adverse effects (17). An association<br />

was observed between this gene and lung cancer (18) and oral cancer (19).<br />

Increased CYP2D6 activity has been related to some malignant processes. In<br />

bladder cancer, Anwar et al, (20) have demonstrated that genetic polymorphism<br />

in CYP2D6 could play an important role as host risk factors for development <strong>of</strong><br />

urinary cancer among Egyptians (20). In north Tunisia, a previous work revealed<br />

that CYP2D6*4 allele did not appear to influence bladder cancer susceptibility<br />

(p > 0.05). A similar result was obtained when he stratified cases group according<br />

to tobacco status (21). However, in other work on a cohort from the middle<br />

centre <strong>of</strong> Tunisia a significant association was found between CYP2D6 (G/G)<br />

wild type and breast carcinoma risk only in postmenopausal patients (p = 0.04)<br />

(22). The data suggest that the increased metabolism <strong>of</strong> one or more agents in<br />

the diet or other environmental agents, mediated by CYP2D6, forms reactive<br />

intermediaries that influence the initiation or promotion <strong>of</strong> cancer in various<br />

tissues (23). The reduced CYP2D6 activity has been related also to greater risk<br />

<strong>of</strong> oral cancer (17).<br />

Other studies demonstrate that patients with lung cancer have shown a greater<br />

frequency <strong>of</strong> extensive metabolizers (EM) genotype. This association is supported<br />

by the discovery that CYP2D6 can activate nitrosamine 4-(methylnitrosamino)-<br />

1-(3-piridyl)-1-butanone, specific to tobacco for reactive metabolites (11). Kato<br />

et al, (24) observed that the levels <strong>of</strong> DNA adducts in the lung were increased<br />

as a function <strong>of</strong> CYP2D6 activity, a result consistent with activation <strong>of</strong> tobacco<br />

mediated by this enzyme (24).<br />

CYP2E1<br />

Many studies have investigated the association between the CYP2E1 5’-flanking<br />

region (RsaI/PstI) polymorphism and head and neck cancer susceptibility, but<br />

the results were conflicting. Using the fixed effects model, Lu et al, (25) have<br />

found significant association between PstI/RsaI polymorphism and head and<br />

neck cancer risk.<br />

Significant results were also found in East Asians and Mix populations when<br />

stratified by ethnicity. However, no significant associations were found for<br />

Caucasians in all genetic models. Stratified analyses according to source <strong>of</strong><br />

controls, significant associations were found only in hospital base controls.<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 41


original article <<br />

In the subgroup analyses by tumor types, significant association was detected<br />

only in oral cancer group, while no significant associations among laryngeal- or<br />

pharyngeal- cancer subgroup. In Egypt, Anwar et al, (20) have demonstrated<br />

any differences in the distribution <strong>of</strong> CYP2E1 polymorphism between bladder<br />

cancer patients and controls as detected by PstI restriction fragment length<br />

polymorphism (RFLP) analysis (20). These previous findings are in accordance<br />

with other work which demonstrates the absence <strong>of</strong> any association between<br />

CYP2E1 gene polymorphisms and breast cancer (22).<br />

CYP2C19<br />

CYP2C19 is one <strong>of</strong> the enzymes involved in the metabolism <strong>of</strong> tamoxifen into<br />

active metabolites. In patients on tamoxifen, CYP2C19*2 and *3 variants,<br />

known for their lack <strong>of</strong> enzyme activity, were associated with a significantly<br />

longer breast cancer survival rate than patients with the wild-type (26). In<br />

Japanese population, a genetic polymorphism <strong>of</strong> CYP2C19 was associated with<br />

susceptibility to biliary tract cancer (27).<br />

Regarding hormone-associated tumors such as ovarian, cervical or prostate<br />

cancers, many conflicting data have been found. In this field, numerous studies<br />

conducted on different populations and ethnic groups have indicated the<br />

absence <strong>of</strong> a major impact <strong>of</strong> the CYP2D6, CYP2C19 and CYP2E1 genes in<br />

cancer risk (28). These previous findings are in accordance with ours which<br />

demonstrate the absence <strong>of</strong> any association between CYP2C19 and CYP2E1<br />

gene polymorphisms with breast cancer. In addition, when compared with the<br />

allele frequencies <strong>of</strong> the CYP2D6, CYP2C19 and the CYP2E1 genes in Italian,<br />

Portuguese and Egyptian populations, we found similar frequencies in healthy<br />

Tunisian individuals (29-31) .<br />

Glutathione S-transferase (GST)<br />

The glutathione S-transferases (GSTs) are key phase II metabolic reaction<br />

enzymes, and they play critical roles in protection against products <strong>of</strong> oxidative<br />

stress and electrophiles. (32). These conjugation reactions facilitate the excretion<br />

<strong>of</strong> many xenobiotics, including carcinogens, toxins, and drugs in the form <strong>of</strong><br />

mercapturic acids. Different GST isozymes have been identified in human<br />

populations, some with tissue-specific expression (33). In mammals they are<br />

expressed at a higher level in the liver, constituting more than 4% <strong>of</strong> total soluble<br />

protein (34). At least five related gene families, mu, alpha, pi, theta and sigma,<br />

have been identified, and genetic polymorphisms have been reported for GSTM1<br />

and GSTT1, resulting in either decreased or altered enzyme activity (35).<br />

GSTM1<br />

Five mu class genes (M1-M5) on chromosome 1p13 have been identified (36).<br />

Inter-individual differences in GSTM1 are due either to gene deletion (the null<br />

genotype) or allelic variation, resulting in production <strong>of</strong> a catalytic active protein<br />

with closely similar catalytic activities which differ by a single base pair in exon<br />

7, introducing a restriction site for Hae II in the gene sequence (37). GSTM1<br />

is expressed at a high level in the liver. Epidemiological studies suggest that<br />

individuals, who are homozygous null, have an increased risk for cancer at a<br />

number <strong>of</strong> sites, lung, bladder, colon, breast (38). The risk <strong>of</strong> lung cancer in<br />

association with GSTM1 null genotype is dependent on the extent <strong>of</strong> tobacco<br />

smoke exposure, the frequency <strong>of</strong> GSTM1 null being significantly higher in the<br />

high exposure group. It may suggest that at low exposure level the genotype<br />

<strong>of</strong> the GSTM1 is less important, and that other detoxification pathways could<br />

handle low doses. Tobacco smoke is considered a risk factor in larynx cancer,<br />

and the GSTM1 null genotype was associated with an increased risk, but only<br />

among smokers that smoke 20 g tobacco/day or less (39).<br />

GSTT1<br />

Two genes have been identified in the theta class: GSTT1 and GSTT2, located<br />

in chromosome 22, in the same region (22q11.2) (40). The null polymorphism<br />

at the GSTT1 shows large variation and occurs at a frequency <strong>of</strong> 16%-40%. The<br />

biological consequences <strong>of</strong> this polymorphism are fairly difficult to predict, as<br />

the enzyme is involved in both detoxification and activation reactions. Lack <strong>of</strong><br />

GSTT1 enzyme activity was observed among 19% <strong>of</strong> Caucasians (41). Many<br />

studies have reported that the GSTT1 null genotype (GSTT1*0) was associated<br />

with increased risk for bladder and lung cancers (42). However, some studies<br />

reported that the risk <strong>of</strong> cancer was increased only among those with the GSTT1<br />

positive (wild-type) genotype (43).<br />

GSTT1, GSTM1 and susceptibility to cancer<br />

Most <strong>of</strong> the studies on the role <strong>of</strong> GST polymorphisms in the development <strong>of</strong><br />

cancer have focused on GSTM1 and GSTT1. In relation to lung cancer, Seidegard<br />

et al, (44) showed that smokers deficient in GSTM1, that is, homozygotes for the<br />

null allele, showed increased risk for this type <strong>of</strong> cancer (44). The presence <strong>of</strong> the<br />

whole gene appeared to protect against chemically-induced cytogenetic damage<br />

and DNA adducts in the lung (45). In the same way, Ryberg et al, (46) showed<br />

that the level <strong>of</strong> DNA adducts in the lungs <strong>of</strong> male smokers was influenced more<br />

by GSTP1 than by GSTM1 (46).<br />

According to these authors, smokers with at least one mutant allele for GSTP1<br />

showed significantly higher levels <strong>of</strong> DNA adducts than controls, while the<br />

GSTM1 null genotype did not show higher levels. When they combined the<br />

two polymorphisms GSTM1 and GSTP1 they observed that patients with the<br />

GSTM1 null genotype and the GSTP1 genotype with at least one mutant allele<br />

had significantly higher levels <strong>of</strong> adducts than other combinations (46).<br />

To-Figueras et al, (47) did not observe a significantly greater frequency <strong>of</strong> the<br />

GSTM1 null genotype in lung cancer cases in a Caucasian population, which<br />

agrees with data from Nyberg et al, (48) (47-48). The frequency <strong>of</strong> the other<br />

genotypes GSTM1 A, GSTM1 B, and GSTM1 A/B did not differ between cases<br />

and controls (Nyberg et al, 1998), unlike the results <strong>of</strong> other studies on bladder<br />

tumors, larynx, and skin, in which a protective role was proposed for GSTM1*A<br />

(49-50) and GSTM1*A/GSTM1*B (51).<br />

Various others authors have described family clustering in oral cancer and the<br />

proposed explanation involves gene polymorphisms for drug-metabolizing<br />

enzymes (52). Jourenkova et al, (53) studied the effect <strong>of</strong> GSTM1 and GSTT1<br />

genotypes on the risk <strong>of</strong> cancer <strong>of</strong> the larynx and observed an increased risk related<br />

to the GSTM1 null genotype and greater risk for GSTT1 null (53). Individuals<br />

lacking both genes GSTM1 and GSTT1 had a two-fold risk, although not<br />

significant, as compared to those with at least one <strong>of</strong> the genes, and a three-fold<br />

risk as compared to those with both genes. In addition, a statistically significant<br />

interaction was observed between the GSTM1 genotypes and levels <strong>of</strong> tobacco<br />

consumption. However, Park et al, (54) failed to find an association between<br />

the GSTM1 null allele and oral cancer (54). Meanwhile, Matthias et al, (55),<br />

studying cancer <strong>of</strong> the upper aero-digestive tract (oral, laryngeal, and pharyngeal<br />

squamous cell carcinoma), did not observe differences in the frequency <strong>of</strong> the<br />

GSTT1 null genotype between cases and controls, but did observe that the<br />

frequency <strong>of</strong> genotype GSTM1 was significantly lower in patients with oral,<br />

laryngeal, and pharyngeal squamous cell carcinoma, suggesting a protective<br />

effect (55).<br />

42 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


Recent epidemiological studies on GSTs and breast cancer have been illustrated.<br />

Some studies suggest an association between the GSTM1 null genotype and<br />

breast cancer in postmenopausal women (56). More recently, Park et al, (54)<br />

observed that the GSTM1 null genotype showed a statistically significant<br />

association with breast cancer, increasing the risk in premenopausal but not in<br />

postmenopausal women, while the GSTT1 null genotype showed similar risk<br />

levels in all the groups analyzed (54). When they combined these two genotypes,<br />

they observed that the presence <strong>of</strong> both null alleles significantly increased the<br />

risk <strong>of</strong> this cancer, especially in premenopausal women and those who consumed<br />

alcohol, suggesting a gene-environment interaction in individual susceptibility<br />

to breast cancer (54). These results disagree with those <strong>of</strong> Curran et al, (57)<br />

who failed to find any association between polymorphisms GSTM1, GSTT1 and<br />

breast cancer in an Australian sample (57). Other independent studies on various<br />

types <strong>of</strong> cancer have also shown this positive association between these genes<br />

and lung cancer (58), bladder cancer (20), colorectal cancer (59), breast cancer<br />

(60), esophageal cancer (61), oral cancer (62), and leukemia (63).<br />

In North Tunisia, we have conducted a case control study to assess the role <strong>of</strong><br />

smoking, slow NAT2 variants, GSTM1 and GSTT1 null genotypes in bladder<br />

cancer development. In all groups <strong>of</strong> patients, we have shown that GSTM1 and<br />

GSTT1 null genotypes did not appear to be a factor affecting bladder cancer<br />

susceptibility. Furthermore, we found that NAT2 slow acetylator individuals<br />

temporarily carrying wild-type GSTT1 or GSTM1 null genotypes have a strong<br />

increased risk <strong>of</strong> bladder cancer (OR=26 and 22.17, respectively) (64).<br />

N-acetyltransferase (NAT)<br />

The NAT locus on human chromosome 8 encodes three distinct NAT genes, the<br />

two active NAT genes, NAT1 and NAT2, separated by an inactive pseudogene.<br />

NAT2 and NAT1 genes are polymorphically expressed, and variation at the<br />

NAT2 locus is responsible for the classic acetylation polymorphism.<br />

NATs catalyze the acetylation <strong>of</strong> carcinogens and other xenobiotics. Arylamines,<br />

hydrazines and hydrazides are metabolized by NAT-mediated N-acetylation.<br />

A majority <strong>of</strong> N-acetylation requires acetyl-CoA as a c<strong>of</strong>actor; however, some<br />

NATs can also utilize N-arylhydroxamic acid as acetyl donors. NATs are<br />

cytosolic enzymes and are found in a large number <strong>of</strong> tissues. The ubiquitous<br />

expression pr<strong>of</strong>ile <strong>of</strong> the NATs suggests a fundamental role in protection against<br />

reactive metabolites. N-acetylation is usually considered a detoxification<br />

process because it renders the nitrogen atom less susceptible to oxidation, an<br />

oxidative process mediated mainly by CYP1A2. NATs can also catalyze the<br />

bioactivation <strong>of</strong> arylhydroxylamines and arylhydroxamic acids by either an<br />

acetyl CoA-dependent O-acetylation or by intramolecular N, O-acetylation<br />

producing reactive N-acetoxyarylamine intermediates. These metabolites are<br />

considered the ultimate carcinogens, because they are able to react with DNA<br />

to form covalent adducts. As a consequence, these enzymes play a role in the<br />

carcinogenesis process initiated by aromatic amines and in arylamine-induced<br />

toxicities. O-acetylation is catalyzed by both NAT1 and NAT2.<br />

Early epidemiological studies have shown that slow acetylators have an<br />

increased risk <strong>of</strong> bladder cancer compared with rapid acetylators (38), especially<br />

in workers occupationally exposed to aromatic amines. However, the NAT2<br />

phenotype did not influence bladder risk among Chinese workers exposed to<br />

benzidine, suggesting that NAT2 N-acetylation is not a critical detoxification<br />

pathway for benzidine. It has been found that mono-functional acetylation is<br />

activation rather than a detoxification pathway for benzidine (65).<br />

NAT2 activity is expressed in human liver and intestinal tissues. The most<br />

studied NAT2 polymorphisms are due to point mutations in the coding region.<br />

Slow acetylators, who possess NAT2 mutant alleles, produce proteins that are<br />

either poorly expressed, unstable, or have partially reduced catalytic activity.<br />

There are at least 15 different NAT2 allelic variants, <strong>of</strong> which the NAT2*4 is<br />

the most predominant and confers a fast acetylating phenotype. Of all the NAT2<br />

allelic variants identified, three (NAT2*5, NAT2*6 and NAT2*7) were shown to<br />

account for most <strong>of</strong> the slow acetylators.<br />

NAT1 protein is found in many human tissues, including the liver and bladder.<br />

NAT1 seems to be primarily responsible for the NAT and O-acetyltransferase<br />

activities in bladder and colon tissues. NAT1 activity in urinary bladder<br />

mucosa represents a major bioactivation step that converts urinary N-hydroxyarylamines<br />

to reactive N-acetoxy esters to form DNA adducts. The activity<br />

<strong>of</strong> NAT1 is significantly correlated with putative aromatic amine adducts.<br />

Individuals classified as slow NAT2 and fast NAT1 would be at the highest risk.<br />

These individuals do also have a higher level <strong>of</strong> carcinogen–DNA adducts in<br />

bladder DNA than people carrying other genotype combinations (66-67). There<br />

are at least 8 identified allelic variants <strong>of</strong> human NAT1. Certain NAT1 variants,<br />

NAT1*10 and NAT1*17, have an elevated N- or O-acetylation compared with<br />

the more common NAT1*4 variant.<br />

A significant association between the NAT1 genotype and lung cancer risk was<br />

observed in smokers, with the slow acetylators having a significantly higher<br />

risk, whereas no association between lung cancer risk and NAT2 was observed<br />

(Bouchardy et al, 1998). However, in a Swedish study, an increased risk for lung<br />

cancer was observed, but only in never-smokers (48).<br />

A slightly increased risk <strong>of</strong> breast cancer was observed in smoking women<br />

classified as NAT2 slow, and this increased risk correlated with a higher<br />

mammary gland DNA adduct level. In contrast, NAT1 did not have any influence<br />

on the adduct level (68).<br />

In case <strong>of</strong> bladder cancer, smokers with the NAT1*10 allele had an increased<br />

risk. There was evidence <strong>of</strong> a gene-dosage effect, as individuals homozygous<br />

for the NAT1*10 alleles had the highest risk. The NAT2 genotype alone did not<br />

significantly influence the bladder cancer risk, but the cancer risk from smoking<br />

exposure was particularly high in those with NAT2 slow alleles in combination<br />

with one or two copies <strong>of</strong> the NAT1*10 allele (69). In Tunisia, For the NAT2<br />

slow acetylator genotype, the NAT2*5/*7 diplotype was found to have a 7-fold<br />

increased risk <strong>of</strong> bladder cancer (OR=7.14) (64). Mechanistically it is assumed<br />

that if an individual is NAT2 slow, arylamines may be rapidly detoxified in the<br />

liver, so that the activated hydroxylated arylamine may never reach the bladder<br />

epithelium, where NAT1 could act upon it.<br />

Occurrence or specific types <strong>of</strong> mutations in oncogenes or tumor suppressor<br />

genes may partially be determined by the NAT2 activity. In bladder cancer, 7 out<br />

<strong>of</strong> 25 acquired mutations in the p53 cancer suppressor gene were transversions,<br />

and 6 out <strong>of</strong> these 7 mutations were in individuals having two slow NAT2 alleles<br />

(70). Differences in metabolic activity play a role in the mutational pattern and<br />

hence the pathobiology <strong>of</strong> the disease. In contrast, no significant association<br />

between p53 gene mutations and NAT2 polymorphisms was observed in patients<br />

with non-small-cell lung cancer, although slow acetylators had an increased risk<br />

compared to fast acetylators (71). Information about the role <strong>of</strong> NAT enzymes<br />

as a risk factor in colorectal cancer is ambiguous (72). In the support <strong>of</strong> NAT as<br />

a risk factor, a Japanese study suggests that people classified as rapid acetylators<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 43


original article <<br />

are more likely to have a point mutation in the K-ras than people classified as<br />

low or intermediate. This suggests that aromatic amines do play a role in the<br />

etiology <strong>of</strong> colon cancer.<br />

Conclusion<br />

Many cancers are caused by various forms <strong>of</strong> environmental and viral exposure<br />

suggests that such causes could be avoided through preventive measures. In<br />

addition, metabolism <strong>of</strong> carcinogens under genetic control as an important<br />

factor in modulating individual susceptibility to cancer is a plausible hypothesis.<br />

Information on susceptibility to cancer is valuable for identifying high-risk<br />

individuals, allowing for early diagnosis and reduction <strong>of</strong> risk exposure to<br />

carcinogens, some <strong>of</strong> them possibly acting as endocrine disruptors. Recent<br />

knowledge on the basic genetics <strong>of</strong> metabolic variation has provided new<br />

possibilities for the study <strong>of</strong> individual susceptibility to cancer induced by the<br />

environmental factors. With the advent <strong>of</strong> techniques based on the polymerase<br />

chain reaction (PCR), it is now possible to identify the genotype <strong>of</strong> an individual<br />

with a series <strong>of</strong> enzymatic polymorphisms involved in the metabolism <strong>of</strong><br />

xenobiotics, some <strong>of</strong> which are potent carcinogens. New molecular biology<br />

techniques have allowed for much more direct correlations between a particular<br />

genotype and the incidence <strong>of</strong> cancer and other chemically-induced diseases.<br />

Given the number <strong>of</strong> polymorphisms, the variability in the expression <strong>of</strong> XMEs,<br />

and the complexity <strong>of</strong> chemical exposure, determination <strong>of</strong> a single polymorphic<br />

enzyme may not be sufficient, and it appears to be necessary to establish a risk<br />

pr<strong>of</strong>ile for each individual or sub-group. The conflictive results observed in the<br />

literature show the still present difficulty to evaluate this complex phenomenon.<br />

Cohorts used for major studies and the number <strong>of</strong> genes responsible for<br />

determining risk are still insufficient and not clear. Some individuals are<br />

more sensitive towards xenobiotics than others, and this may in part be due to<br />

differences in metabolic capacity. A genetic high risk pr<strong>of</strong>ile cannot be made<br />

at the present time, as the pr<strong>of</strong>ile will depend not only on the compound under<br />

investigation but also on the adverse health effect. Studies combining various<br />

XME genotypes from phases I and II <strong>of</strong> metabolism may be provide more<br />

information than the analysis <strong>of</strong> individual genes, since if genetic susceptibility<br />

is partially mediated by polymorphic variation, the risk associated with only one<br />

locus is probably small, due to the multiplicative interaction model probably at<br />

play. One <strong>of</strong> the futture challenges in molecular epidemiology may be resides in<br />

the ability to evaluate different scenarios in which interactions among several<br />

genetic polymorphisms, and among gene/s and environmental carcinogens yield<br />

different susceptibility levels on cancer etiology.<br />

References<br />

1. Parkinson A (1996). Biotransformation <strong>of</strong> xenobiotics in: casarett and<br />

doull’s toxicology. The Basic Science <strong>of</strong> Poisons, 6: 113-186.<br />

2. Doll R and Peto R (1981). The causes <strong>of</strong> cancer: quantitative estimates <strong>of</strong><br />

avoidable risks <strong>of</strong> cancer in the United States today. <strong>Journal</strong> <strong>of</strong> the National<br />

Cancer Institute, 66: 1191-1308.<br />

3. Raunio H, Husgafvel-pursiainen K, Anttila S, Hietanen E, Hirvönen A and<br />

Pelkonen O (1995). Diagnosis <strong>of</strong> polymorphisms in carcinogen-activating and<br />

inactivating enzymes and cancer susceptibility. A review gene, 159: 113-121.<br />

4. Kawajiri K, Nakachi K, Imai K, Yoshii A, Shinoda N and Watanabe J<br />

(1990). Identification <strong>of</strong> genetically high risk individuals to lung cancer by<br />

dna polymorphisms <strong>of</strong> the cytochrome gene. Febs Letters, 263: 131-133.<br />

5. Ingelman-sundberg M (1998). Functional consequences <strong>of</strong> polymorphism<br />

<strong>of</strong> xenobiotic metabolising enzymes. Toxicology Letters, 102-103:155-160.<br />

6. Perera FP (1997). Environment and cancer: who are susceptible? Science,<br />

278: 1068-1073.<br />

7. Nebert DW (1991). Role <strong>of</strong> genetics and drug metabolism in human cancer<br />

risk. Mutation Research, 247: 267-281.<br />

8. Kawajiri K, Nakachi K, Imai K, Watanabe J, Hayashi S (1993). The cyp1a1<br />

gene and cancer susceptibility. Critical Reviews in hematologic oncology,<br />

14: 77-87.<br />

9. Autrup H (2000). Genetic polymorphisms in human xenobiotic metabolizing<br />

enzymes as susceptibility factors in toxic response. Mutation research, 464:<br />

65-76.<br />

10. Gough AC, Smith D, Howell, Wolf R, Bryant P and Spurr K (1993).<br />

Localization <strong>of</strong> the cyp2d gene locus to human chromosome 22q131 by<br />

polymerase chain reaction in situ hybridization and linkage. Analysis<br />

Genomics, 15: 430-432.<br />

11. Crespi L, Penman B, Gelboin V, and Gonzalez J (1991). A tobacco smokederived<br />

nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone is<br />

activated by multiple human cytochrome p-450s including the polymorphic<br />

human cytochrome p-4502d6. Carcinogenesis, 12: 1197-1201.<br />

12. Bertilsson L (1995). Geographical/interracial differences in polymorphic<br />

drug oxidation current state <strong>of</strong> knowledge <strong>of</strong> cytochromes p450 (cyp) 2d6<br />

and 2c19. Clinical Pharmacokinet, 29: 192-209.<br />

13. Idle R, Mahgoub A, Lancaster R and Smith L (1978). Hypotensive response<br />

to debrisoquine and hydroxylation phenotype. Life Sciences, 22: 979-984.<br />

14. Nebert W and Gonzalez J (1987). P450 genes: structure evolution and<br />

regulation. Annual Review <strong>of</strong> Biochemistry, 56: 945-993.<br />

15. Pavanello S and Clonfero E (2000). Biological indicators <strong>of</strong> genotoxic<br />

risk and metabolic polymorphisms. Mutation research/reviews in mutation<br />

research, 463: 285-308.<br />

16. Sachse C, Brockmöller J, Bauer S and Roots I (1997). Cytochrome p450<br />

2d6 variants in a caucasian population: allele frequencies and phenotypic<br />

consequences. American <strong>Journal</strong> <strong>of</strong> Human Genetics, 60: 284-295.<br />

17. Chen H, Sandler P, Taylor A, Shore L, Liu E, Bloomfield D and Bell A<br />

(1996). Increased risk for myelodysplastic syndromes in individuals with<br />

glutathione transferase theta 1 (gstt1). Gene Defect Lancet, 347: 295-297.<br />

18. Roots I, Drakoulis N, Ploch M, Heinmeyer G, Loddenkemper R, Minks T,<br />

Nitz M, Otte f and Koch M (1988). Debrisoquine hydroxylation phenotype<br />

acetylation phenotype and abo blood groups as genetic host factors <strong>of</strong> lung<br />

cancer risk. Klinische wochenschrift, 66: 87-97.<br />

19. Worrall S, Corrigan M, High A, Starr D, Matthias C, Wolf R, Jones W, Hand<br />

P, Gilford J, Farrell W, Hoban P, Fryer A and strange R (1998). Susceptibility<br />

and outcome in oral cancer: preliminary data showing an association with<br />

polymorphism in cytochrome p450 cyp2d6. Pharmacogenetics, 8: 433-439.<br />

20. Anwar W, Abdel-rahman S, El-zein A, Mostafa M, and Au W (1996).<br />

Genetic polymorphism <strong>of</strong> gstm1 cyp2e1 and cyp2d6 in Egyptian bladder<br />

cancer patients. Carcinogenesis, 17: 1923-1929.<br />

21. Ouerhani S, Marrakchi R, Bouhaha R, Ben Slama M, Sfaxi M, Ayed M,<br />

Chebil M and Elgaaied A (2008). The role <strong>of</strong> cyp2d6*4 variant in bladder<br />

cancer susceptibility in tunisian patients. Bull Cancer, 95: 1-4.<br />

22. Khedhaier A, Hassen E, Bouaouina N, Gabbouj S, Ahmed S, Chouchane L<br />

(2008). Implication <strong>of</strong> Xenobiotic Metabolizing Enzyme gene (CYP2E1,<br />

CYP2C19, CYP2D6, mEH and NAT2) polymorphisms in breast carcinoma.<br />

BMC Cancer, 8: 109.<br />

23. Nebert W (1997). Polymorphisms in drug-metabolizing enzymes: what is<br />

their clinical relevance and why do they exist? American <strong>Journal</strong> <strong>of</strong> Human<br />

44 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


Genetics, 60: 265-271.<br />

24. Kato S, Bowman ED, Harrington AM, Blomeke B and Shields PG<br />

(1995). Human lung carcinogen-DNA adduct levels mediated by genetic<br />

polymorphisms in vivo. <strong>Journal</strong> <strong>of</strong> the National Cancer Institute, 87: 902-<br />

907.<br />

25. Lu D, Yu X and Du Y (2010). Meta-analyses <strong>of</strong> the effect <strong>of</strong> cytochrome<br />

P450 2E1 gene polymorphism on the risk <strong>of</strong> head and neck cancer. Mol Biol<br />

Rep (In press).<br />

26. Ruiter R, Bijl MJ, Van Schaik RH, Berns EM, H<strong>of</strong>man A, Coebergh JW, Van<br />

Noord C, Visser LE and Stricker BH (2010). CYP2C19*2 polymorphism is<br />

associated with increased survival in breast cancer patients using tamoxifen.<br />

Pharmacogenomics, 11: 1367-1375.<br />

27. Isomura Y, Yamaji Y, Ohta M, Seto M, Asaoka Y, Tanaka Y, Sasaki T, Nakai<br />

Y, Sasahira, N, Isayama H, Tada M, Yoshida H, Kawabe T (2010). A genetic<br />

polymorphism <strong>of</strong> CYP2C19 is associated with susceptibility to biliary tract<br />

cancer. J Gastroenterol, 45: 1045-1052.<br />

28. Agundez JA (2004). Cytochrome P450 gene polymorphism and cancer.<br />

Curr Drug Metab, 5: 211-24.<br />

29. Lemos, MC, Carrilho F, Rodrigues F., Coutinho E, Gomes L, Carvalheiro<br />

M, Regateiro FJ (2007). Genetic polymorphism <strong>of</strong> CYP2D6 influences<br />

susceptibility to papillary thyroid cancer. Clin Endocrinol, 67: 180-3.<br />

30. Ruas JL, Lechner MC (1997). Allele frequency <strong>of</strong> CYP2C19 in a Portuguese<br />

population. Pharmacogenetics, 7: 333-5.<br />

31. Hamdy SI, Hiratsuka M, Narahara K, El-Enany M., Moursi N, Ahmed MS<br />

(2002). Allele and genotype frequencies <strong>of</strong> polymorphic cytochromes P450<br />

(CYP2C9 CYP2C19 CYP2E1) and dihydropyrimidine dehydrogenase<br />

(DPYD) in the Egyptian population. Br J Clin Pharmacol, 53: 596-603.<br />

32. Pickett B and Lu H (1989). Glutathione s-transferases: gene structure<br />

regulation and biological function. Annual Review <strong>of</strong> Biochemistry, 58: 743-<br />

764.<br />

33. Suzuki T, Coggan M, Shaw D and Board G (1987). Electrophoretic and<br />

immunological analysis <strong>of</strong> human glutathione s-transferase isozymes.<br />

Annals <strong>of</strong> Human Genetics, 51: 95-106.<br />

34. Eaton L and Bammler K (1999). Concise review <strong>of</strong> the glutathione<br />

s-transferase and their significance to toxicology. Toxicology Science, 49:<br />

156-164.<br />

35. Landi, S (2000). Mammalian class theta gst and differential susceptibility to<br />

carcinogens: a review. Mutation Research, 463: 247-283.<br />

36. Pearson W, Vorachek W, Xu S, Berger R, Hart I, Vannais D and Patterson<br />

D (1993). Identification <strong>of</strong> class-mu glutathione transferase genes gstm1gstm5<br />

on human chromosome 1p13. American <strong>Journal</strong> <strong>of</strong> Human Genetics,<br />

53: 220-223.<br />

37. Fryer A, Zhao L, Alldersea J, Pearson R and Strange R (1993). Use <strong>of</strong> sitedirected<br />

mutagenesis <strong>of</strong> allele-specific pcr primers to identify the gstm1<br />

a gstm1 b gstm1 ab and gstm1 null polymorphisms at the glutathione<br />

s-transferase gstm1 locus. Biochemistry <strong>Journal</strong>, 295: 313-315.<br />

38. D’Errico A, Taioli X, Chen X, and Vineis P (1996). Genetic metabolic<br />

polymorphisms and the risk <strong>of</strong> cancer: a review <strong>of</strong> the literature. Biomarkers,<br />

1: 149–173.<br />

39. Jourenkova-mironova N, Wikman H, Bouchardy C, Yoho A, Dayer P<br />

(1998). Role <strong>of</strong> glutathione s-transferase gstm1 gstm3 gstp1 and gstt1<br />

genotypes in modulating susceptibility to smoking-related lung cancer.<br />

Pharmacogenetics, 8: 495-502.<br />

40. Webb G, Vaska V, Coggan M and Board P (1996). Chromosomal<br />

localization <strong>of</strong> the gene for the human theta class glutathione transferase<br />

(gstt1). Genomics, 33: 121-123.<br />

41. Garte S, Gaspari L, Alexandrie AK, Ambrosone C, Autrup H, Autrup JL,<br />

Baranova H., Bathum L, Benhamou S, B<strong>of</strong>fetta P (2001). Metabolic gene<br />

polymorphism frequencies in control populations. Cancer Epidemiol<br />

Biomarkers Prev, 10: 1239-48.<br />

42. Abdel-Rahman SZ, Anwar WA, Abdel-Ala WE, Mostafa HM and Au WW<br />

(1998). GSTM1 and GSTT1 genes are potential risk modifiers for bladder<br />

cancer. Cancer Detect Prev, 22: 129-38.<br />

43. Hengstler JG, Kett A, Arand M, Oesch-Bartlomowicz B, Oesch F, Pilch<br />

H, Tanner B (1998). Glutathione S-transferase T1 and M1 gene defects in<br />

ovarian carcinoma. Cancer Lett, 130: 43-8.<br />

44. Seidegard J, Vorachek R, Pero W and Pearson W (1988). Hereditary<br />

differences in the expression <strong>of</strong> the human glutathione transferase active on<br />

trans-stilbene oxide are due to a gene deletion. Proceedings <strong>of</strong> the National<br />

Academy <strong>of</strong> Sciences <strong>of</strong> the USA, 85: 7293-7297.<br />

45. Scarpato R, Hirvönen A, Migliore L, Flack G and Norppa H (1997).<br />

Influence <strong>of</strong> gstm1 and gstt1 polymorphisms on the frequency <strong>of</strong><br />

chromosome aberrations in smokers and pesticide-exposed greenhouse<br />

workers. Mutation Research, 389: 227-235.<br />

46. Ryberg D, Skaug V, Hewer A, Phillips H, Harries W, Wolf R (1997).<br />

Genotypes <strong>of</strong> glutathione transferase m1 and p1 and significance for lung<br />

DNA adduct levels and cancer risk. Carcinogenesis, 18: 1285-1289.<br />

47. To-figueras J, Gene M, Gomez-catalan J, Galan C, Fuentes M, Ramon M,<br />

Rrodamilans M, Huguet E and Corbella J (1997). Glutathione s-transferase<br />

m1 (gstm1) and t1 (gstt1) polymorphisms and lung cancer risk among<br />

northwestern mediterraneans. Carcinogenesis, 18: 1529-1533.<br />

48. Nyberg SM, Hou K, Hemminki B, and Pershagen G (1998). Glutathione<br />

S-transferase M1 and N-acetyltransferase 2 genetic polymorphisms and<br />

exposure to tobacco smoke in nonsmoking and smoking lung cancer patients<br />

and population controls. Cancer Epidemiol Biomarkers Prev, 7: 875–883.<br />

49. Brockmöller J, Cascorbi I, Kerb R, & Roots I (1996). Combined analysis<br />

<strong>of</strong> inherited polymorphism in arylamine n-acetyltransferase 2 glutathione<br />

s-transferases m1 and t1 microsomal epoxide hydrolase and cytochrome<br />

p450 enzymes as modulators <strong>of</strong> bladder cancer risk. Cancer research, 56:<br />

3915-3925.<br />

50. Brockmoller J, Kaiser R, Kerb I, Cascorbi V, Jaeger V, and Roots I (1996).<br />

Polymorphic enzymes <strong>of</strong> xenobiotic metabolism as modulators <strong>of</strong> acquired<br />

p53 mutations in bladder cancer. Pharmacogenetics, 6: 535–545.<br />

51. Jahnke V, Strange R, Matthias C and Fryer A (1996). Glutathione<br />

s-transferase and cytochrome-p-450 polymorphism as risk factors for<br />

squamous cell carcinoma <strong>of</strong> the larynx. American journal <strong>of</strong> surgery, 172:<br />

671-673.<br />

52. Foulkes W, Brunet S, Kowalski P, Narod and Franco L (1995). Family<br />

history <strong>of</strong> cancer is a risk factor for squamous cell carcinoma <strong>of</strong> the head<br />

and neck in Brazil: a case-control study. International <strong>Journal</strong> <strong>of</strong> Cancer,<br />

63: 769-773.<br />

53. Jourenkova N, Reinikainen M, Bouchardy C, Dayer P, Benhamou S and<br />

Hirvönen A (1998). Larynx cancer risk in relation to glutathione s-transferase<br />

m1 and t1 genotypes and tobacco smoking. Cancer Epidemiology<br />

Biomarkers and Prevention, 7: 19-23.<br />

54. Park J, Muscat E, Ren Q, Schantz S, Harwick D, Stern C, Pike V, Richie P<br />

and Lazarus P (1997). Cyp1a1 and gstm1 polymorphisms and oral cancer<br />

risk. Cancer Epidemiology Biomarkers and prevention,6: 791-797.<br />

55. Matthias C, Bockmühl U, Jahnke V, Petersen I, Dietel M, Fryer A, Strange<br />

RC.(1998).Effect <strong>of</strong> gene polymorphism on detoxifying glutathione-Stransferase<br />

enzymes on chromosomal stability <strong>of</strong> squamous epithelial<br />

carcinomas in the area <strong>of</strong> the head-neck. Laryngorhinootologie, 77:201-6.<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 45


original article <<br />

56. Charrier J, Maugard C, Mevel B and Bignon J (1999). Allelotype influence<br />

at glutathione s-transferase m1 locus on breast cancer susceptibility. British<br />

<strong>Journal</strong> <strong>of</strong> cancer ,79: 346-353.<br />

57. Curran J, Weinstein R and Griffiths R (2000). Polymorphism <strong>of</strong> glutathione<br />

s-transferase (gstm1 gstp1 and gstt1) and breast cancer susceptibility.<br />

Cancer Letters, 153: 113-120.<br />

58. Lan Q, He X, Costa J, Tian L, Rothman H, Mumford J (2000). Indoor coal<br />

combustion emissions gstm1 and gstt1 genotypes and lung cancer risk: a<br />

case-control study in xuan wei china. Cancer Epidemiology Biomarkers and<br />

Prevention, 9: 605-608.<br />

59. Deakin M, Elder J, Hendrickse C, Peckham D, Baldwin D, <strong>Pan</strong>tin C,<br />

Fryer A and Strange R (1996). Glutathione s-transferase gstt1 genotypes<br />

and susceptibility to cancer: studies <strong>of</strong> interactions with gstm1 in lung oral<br />

gastric and colorectal cancers. Carcinogenesis, 17: 881-884.<br />

60. Mitrunen K, Jourenkova N, Kataja V, Eskelinen M, Kosma M, Benhamou<br />

S, Vainio H, Uusitupa M (2001). Glutathione s-transferase m1 m3 p1<br />

and t1 genetic polymorphisms and susceptibility to breast cancer. Cancer<br />

Epidemiology Biomarkers and Prevention, 10: 229-236.<br />

61. Van lieshout E, Roel<strong>of</strong>s M, Dekker S, Mulder J, Wobbes T, Jansen B and<br />

Peters H (1999). Polymorphic expression <strong>of</strong> the glutathione s-transferase p1<br />

gene and its susceptibility to barrett’s esophagus and esophageal carcinoma.<br />

Cancer Res, 59: 586-589.<br />

62. Nair U, Matthew B and Bartsch H (1999). Glutathione s-transferase m1 and<br />

t1 null genotypes as risk factors for oral leukoplakia in ethnic indian betel<br />

quid/tobacco chewers. Carcinogenesis, 20: 743-748.<br />

63. Chen C, Liu G, Pui C, Rivera K, Sandlund T, Ribeiro R (1997). Higher<br />

frequency <strong>of</strong> glutathione s-transferase deletions in black children with acute<br />

lymphoblastic. Leukemia blood, 89: 1701-1707.<br />

64. Kamel R, Slah O, Raja M, Mohamed R, Mohamed S, Mohsen A, Mohamed<br />

C, Amel G (2009). Combined effect <strong>of</strong> smoking and inherited polymorphisms<br />

in arylamine N-acetyltransferase 2 glutathione S-transferases M1 and T1 on<br />

bladder cancer in a Tunisian population. Cancer Genetics and Cytogenetics,<br />

190: 101-107.<br />

65. Rothman N, Bhatnagar VK, Hayes R, Zenser TV (1996). The impact <strong>of</strong><br />

interindividual variation in NAT2 activity on benzidine urinary metabolites<br />

and urothelial DNA adducts in exposed workers. Proc Natl Acad Sci USA,<br />

93: 5084–5089.<br />

66. Badawi AF, Hirvonen A, Bell DA, Lang NP, and Kadlubar FF (1995). Role<br />

<strong>of</strong> aromatic amine acetyltransferases NAT1 and NAT2 in carcinogen–DNA<br />

adduct formation in the human urinary bladder. Cancer Res, 55: 5230–5237.<br />

67. Badawi AF, Stern SJ, Lang NP, and Kadlubar FF (1996). Cytochrome P-450<br />

and acetyltransferase expression as biomarkers <strong>of</strong> carcinogen–DNA adduct<br />

levels and human cancer susceptibility. Prog Clin Biol Res, 395: 109–140.<br />

68. Pfau EM, Stone U, Brockstedt PL, Carmichael H, Phillips DH (1998).<br />

DNA adducts in human breast tissue: association with N-acetyltransferase-2<br />

(NAT2) and NAT1 genotypes. Cancer Epidemiol Biomarkers Prev, 7:<br />

1019–1025.<br />

69. Taylor JA, Umbach DM, Stephens E, Castranio T (1998). The role <strong>of</strong><br />

N-acetylation polymorphisms in smoking-associated bladder cancer:<br />

evidence <strong>of</strong> a gene-gene-exposure three-way interaction. Cancer Res, 58:<br />

3603–3610.<br />

70. Brockmöller J, Kerb R, Drakoulis N, Staffeldt B (1994). Glutathione<br />

s-transferase m1 and its variants a and b as host factors <strong>of</strong> bladder cancer<br />

susceptibility: a case-control study. Cancer research, 54: 4103-4111.<br />

71. Oyama T, Kawamoto T, Mizoue T, Yasumoto K, Kodama Y and Mitsudomi<br />

T (1997). N-acetylation polymorphism in patients with lung cancer and its<br />

association with p53 gene mutation. Anticancer Res, 17: 577–581.<br />

72. Oda Y, Tanaka M and Nakanishi I (1994). Relation between the<br />

occurrence <strong>of</strong> K-ras gene point mutations and genotypes <strong>of</strong> polymorphic<br />

N-acetyltransferase in human colorectal carcinoma. Carcinogenesis,<br />

15:1365–1369.<br />

46 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


notes <<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 47


original article <<br />

Hyp<strong>of</strong>ractionated radiotherapy versus conventional radiotherapy in treatment <strong>of</strong><br />

glioblastoma multiforme<br />

Mohamed Abdelgawad, MD 1 , Eman Ismail MD 1 , Nashwa Nawar, MD 1<br />

(1) Clinical oncology department, Faculty <strong>of</strong> Medicine, Zagazig University, Egypt<br />

Corresponding author: Mohamed Abdelgawad, MD<br />

Clinical oncology department, Faculty <strong>of</strong> Medicine, Zagazig University, Egypt<br />

E-mail: gawad2@yahoo.com<br />

ISSN: 2070-254X<br />

Abstract<br />

Purpose : This study was planned to evaluate the safety and efficacy <strong>of</strong><br />

hyp<strong>of</strong>ractionated radiotherapy(HRT) in treatment <strong>of</strong> glioblastoma multiforme<br />

(GM).<br />

Patients and methods : Twenty adult patients with (GM) were prospectively<br />

treated with (HRT) after surgical excision. HRT was given 3 days a week with a<br />

tumor dose <strong>of</strong> 3Gy/fraction. There were two phases <strong>of</strong> treatment, the first phase<br />

was 12 fractions, and the second phase was three fractions with smaller field<br />

size. The total dose was 50 Gy/15 fraction. The results were compared with a<br />

control group retrospectively treated with conventional radiotherapy.<br />

Results : Twenty patients with GM were treated. Excisional biopsy was done in<br />

15% <strong>of</strong> cases , 25% <strong>of</strong> cases underwent subtotal excision while 60% <strong>of</strong> cases<br />

were operated with total excision. As regard historical group 20% <strong>of</strong> cases<br />

underwent excisional biopsy ,subtotal excision in 30% and total excision in 50%<br />

<strong>of</strong> cases . The study revealed that median overall survival 6.5 months and 6<br />

months for (HRT) and conventional group respectively. The progression free<br />

survival was 6 months and 5 months for the (HRT) and conyentional group<br />

respectively. Treatment was well tolerated with minimal acute toxicities.<br />

Conclusion: Although HRT didn’t improve overall survival or progression free<br />

survival in GM compared with conventional group but the treatment duration<br />

was reduced which may be <strong>of</strong> palliative benefit in such group <strong>of</strong> patients. Further<br />

studies could be useful to determine the optimal fraction size for GM when HRT<br />

is used as an adjuvant treatment.<br />

Introduction<br />

Glioblastoma multiforme is one <strong>of</strong> the most common primary brain tumors in<br />

adults(1). Half <strong>of</strong> the brain tumors in adults are GM(2). The incidence <strong>of</strong> this<br />

disease increases after the age <strong>of</strong> forty (3).<br />

Although brain tumors are uncommon in comparison with other cancers such<br />

as breast or lung cancer. They exert a tremendous toll upon the patients, their<br />

spouses and families because <strong>of</strong> the sever neurologic disability they produce.<br />

The incidence and mortality from primary brain tumors appears to be rising,<br />

particularly amongst the elderly(4).<br />

High grade primary brain tumors such as anaplastic astrocytoma and glioblastoma<br />

are feard because <strong>of</strong> their aggressive course. In spite <strong>of</strong> intensive therapy with<br />

surgical resection, radiotherapy and chemotherapy the prognosis for malignant<br />

gliomas remains poor, with median survival <strong>of</strong> one year(5).The improvement in<br />

diagnostic and treatment modalities in the last years doesn’t affect the overall<br />

prognosis and natural course <strong>of</strong> GM(1).<br />

Although GM carries a fatal prognosis, postoperative radiotherapy has been<br />

shown to increase the median survival compared with that for patients treated<br />

with surgery alone(6) .<br />

The standard dose <strong>of</strong> radiation given after surgical resection is 60 Gy delivered<br />

in 1.8 – 2.0 Gy / fration. Dose escalation through standard fractionation to 70 –<br />

90 Gy has recently been attempted with conformal techniques, although changes<br />

in the pattern <strong>of</strong> failure have been observed, survival improvement hasn’t been<br />

achieved(7) .<br />

Hyperfractionation (giving a smaller fraction size twice daily) to a total dose 72<br />

Gy has shown no specific benefit for GM(8) .<br />

From the radiobiologic standpoint, late responding tissues such as neural tissue<br />

should be more responsive to fewer, but larger dose fractions <strong>of</strong> radiation,<br />

therefore to control CNS tumors such as GM adequately, it is likely that the<br />

radiation dose given must exceed the tolerance <strong>of</strong> the surrounding brain tissue,<br />

resulting in an unacceptable side effect pr<strong>of</strong>ile. However, this basic problem may<br />

be overcome if the tumor alone is treated to these higher doses, and the normal<br />

tissue is spared(9).<br />

Several prospective trials have been performed using hyp<strong>of</strong>ractionated radiation<br />

dosing for the treatment <strong>of</strong> primary GM(9). A randomized study from Maria<br />

Slodowska – Curie Memorial Center, Poland evaluated 44 patients with GM<br />

who were treated with three split courses <strong>of</strong> hyp<strong>of</strong>rationated radiotherapy to a<br />

total dose <strong>of</strong> 50 Gy and such study gave a statistically significant 2-year survival<br />

benefit compared with conventional therapy(10).<br />

In Royal Marsden Hospital, a phase I – II study had been performed using<br />

5-Gy fractions <strong>of</strong> stereotactic RT to 20 – 50 Gy proved to be efficacious and<br />

tolerable(11).<br />

48 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


In 1997 Arslan and his colleagues designed a prospective study to evaluate the<br />

safty and efficacy <strong>of</strong> postoperative HRT in GM patients in terms <strong>of</strong> overall and<br />

progression – free survival, treatment was well tolerated, acute toxicity was<br />

minimal and the study supported that HRT could be used instead <strong>of</strong> conventional<br />

and hyperfractionated radiotherapy(12).<br />

A non randomized study involved 30 patients treated with a hyp<strong>of</strong>ractionated<br />

course <strong>of</strong> 42 Gy in 14 fractions. The median survival and toxicity were<br />

comparable to that after standard fractionation(13).<br />

Although the methods <strong>of</strong> such trials varied as regard fraction size, total dose,<br />

and the overall treatment time, all were shown to have an acceptable toxicity and<br />

encouraging results.<br />

The aim <strong>of</strong> this study was to evaluate the safety and efficacy <strong>of</strong> adjuvant<br />

hyp<strong>of</strong>ractionated radiotherapy for patients with GM<br />

Patients and Methods<br />

From December 2007 to January 2009,twenty adult patients diagnosed<br />

histologically as GM were referred to clinical oncology department Zagazige<br />

University Hospitals. These cases were enrolled in group A which were treated<br />

with hyp<strong>of</strong>ractionated radiotherapy and were compared with a historical<br />

control group treated with conventional radiotherapy. Table(1)shows patient<br />

characteristics .<br />

The types <strong>of</strong> operation which were performed for the study group and the control<br />

group ,respectively, were : total tumor excision 12 (60%) and 10 (50%); subtotal<br />

excision 5 (25%) and 6 (30%); biopsy 3 (15%) and 4 (20%) .<br />

The diagnosis was confirmed to be GM by the pathologic examination <strong>of</strong> the<br />

surgical specimen. After wound healing; on the average 3 weeks postoperatively;<br />

HRT was delivered with linear accelerators . The irradiated volume was<br />

determined by preoperative compeutrized tomography or the magnetic resonance<br />

imaging. The gross target volume (GTV) for both the initial volume (GTV1) and<br />

the conedown volume(GTV2) is obtained from the MRI. GTV1 includes the<br />

contrast enhancing lesion, the surgical resection cavity and surrounding edema; a<br />

2.0-cm margin is added to form PTV1. GTV2 for the conedown treatment should<br />

include the contrast enhancing lesion (without edema) plus a 2.5-cm margin to<br />

form PTV2. In the first phase <strong>of</strong> treatment, the tumor and edema around were<br />

irradiated with 2-3cm margin from the normal brain tissue. In the second phase<br />

the tumor region only was irradiated.<br />

The HRT was applied 3 days a week with a tumor dose 3.33 Gy per fraction. At<br />

the first phase <strong>of</strong> treatment 12 fractions and at the second phase 3 fractions with<br />

smaller fields were delivered. The total dose was 50 Gy/15 fraction 15 weeks.<br />

The histortical control group were treated with conventional radiotherapy with<br />

total dose 60 Gy in 30 fractions over 6 weeks one year overall survival and<br />

progression free survival times were calculated from date <strong>of</strong> operation using the<br />

Kaplan meier method.<br />

Daily dexamethasone 16 mg I.V was given as a brain dehydrating<br />

measure to all patients during HRT. After HRT 50% <strong>of</strong> patients continued<br />

maintenance dexamethasons 4 mg daily for about 6 weeks with gradual<br />

discontinuation also antiepileptic treatment was given continuously.<br />

After Radiotherapy completed, patients were evaluated monthly by physical<br />

examination, neurologic examination and determination <strong>of</strong> performance status.<br />

Radiological examination by C.T or MRI were done every three months.<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 49<br />

Results<br />

Twenty patients were included in the study group, 12 males (60%) and 8<br />

females (40%) the mean age was 55 years (rang 40 – 72). All patients had an<br />

initial Karn<strong>of</strong>sky performance status > 70 . 75% <strong>of</strong> the study group had tumor<br />

dimension 4 cm or less all patients had supratentorial tumor. Sixteen patients<br />

(80%) presented with symptoms <strong>of</strong> increased intracranial tension i.e headache,<br />

nausea, vomiting, mental changes and blurring <strong>of</strong> vision. Forty percent <strong>of</strong> the<br />

patients had hemiparesis–hypoesthesia and 30% had seizures.The mean duration<br />

<strong>of</strong> illness was 10 weeks (range 2 – 72).<br />

The patients <strong>of</strong> the study group were irradiated with HRT after the operation.<br />

Treatment was well tolerate and acute toxicities were mild, serious acute side<br />

affect were not observed. In all cases, radiation necrosis as identified on MRI<br />

appeared to be within the high does area which determined during treatment<br />

planning.<br />

All twenty patients were followed up regularly until death the overall survival<br />

and time to disease progression were measured from the day <strong>of</strong> radiotherapy<br />

completion.<br />

The median overall survival <strong>of</strong> the study group was 6.5 months (range1-14) and<br />

the median <strong>of</strong> progression free survival was 6 months (range 1-15).<br />

As regard the historical conventional group the median overall survival was 6<br />

months (range 2-18) while the progression free survival was 5 months (range<br />

2-14) Table(2) fig 1,2.<br />

Discussion<br />

Although prognosis <strong>of</strong> GM is ominous, some characteristics <strong>of</strong> the patients,<br />

tumor and treatment can affect the natural course <strong>of</strong> this neoplasm. Known<br />

and reported prognostic factors related to patients and tumor characteristics are<br />

patients age, performance status before operation, tumor size, tumor localization<br />

and symptoms <strong>of</strong> their duration.The treatment <strong>of</strong> glioblastoma is a serious<br />

clinical problem. If no adjuvant treatment is given after operation, the median<br />

survival time is about 4 months(1). Surgery plus post operative radiotherapy is<br />

standard and optimal treatment (14).<br />

With respect to survival duration, the optimum dose <strong>of</strong> radiation defined in<br />

randomized studies is 60 Gy in 30 fractions given over 6 weeks (15). The dose<br />

per fraction is between 1.8 – 2 Gy (14) .<br />

A meaningful improvement in survival from doses above 60 Gy has not been<br />

demonstrated (16). Postoperative HRT has been tested from the radiobiologic<br />

stand point, GM are relatively radioresistant and respond more like neural tissue,<br />

which is a late responding tissue the a/B ratio is small for late responding tissues<br />

this implies that a late responding tissue should be more sensitive to fewer,<br />

higher dose fractions (HRT) (9).Postoperative HRT has been tested in some trials<br />

(12). Hyp<strong>of</strong>ractionated radiotherapy has three advantages over standard therapy.


original article <<br />

First, fewer overall treatment session are needed and the total treatment time is<br />

shorter. With a uniformly fatal disease such as GM this is an important concern<br />

in terms <strong>of</strong> quality <strong>of</strong> life owing to the significant time commitment required<br />

to undergo RT. This regimen <strong>of</strong>fers a treatment course that is completed in 2,<br />

instead <strong>of</strong> 6 weeks which may be preferable in certain subsets <strong>of</strong> patients such<br />

as those with poor performance status unresectable disease when palliation is<br />

the primary treatment goal. Second, the reduced number <strong>of</strong> treatment sessions<br />

confers a cost advantage over standard therapy. Finally radiobiologic advantage<br />

to hyp<strong>of</strong>ractionation with GM my exist (9). The general consensus is that<br />

conventional intensive treatment which carries morbidity may not be appropriate<br />

for patients with adverse prognostic features who may have a limited gain in<br />

life expectancy. These patients tend to be <strong>of</strong>fered either supportive care alone or<br />

short palliative radiotherapy with the aim <strong>of</strong> avoiding both prolonged treatment<br />

which would take up a large part <strong>of</strong> remaining life and prolonged side effects <strong>of</strong><br />

intensive radiotherapy (17).<br />

Slotman etal (18) treated thirty patients with HRT in the form <strong>of</strong> 42 Gy given<br />

in 3 weeks (5 daily fractions <strong>of</strong> 3 Gy each per week) the median follow up time<br />

was 24 months and 80% <strong>of</strong> patients recurred after 6.5 months, Age performance<br />

status and type <strong>of</strong> operation were found as prognostic factors.<br />

Such study reported that the results <strong>of</strong> HRT were similar with conventional<br />

radiotherapy. None <strong>of</strong> the patients experienced sever acute or late complications.<br />

Glinski et al (10) compared 44 patients treated with HRT or conventional<br />

radiotherapy in their randomized prospective trial. Two – year survival was 10%<br />

in the conventional arm and 23% in the HRT arm. Treatment in both arms was<br />

well tolerated.<br />

Lang et al (19) treated 38 patients, diagnosed as glioblastoma multiforme with<br />

HRT where 3.5 Gy per fraction were given with 5 fractions per week up to total<br />

dose <strong>of</strong> 42 Gy. Median survival was 11.5 months (45.7 weeks; rang 29.6 – 63.6).<br />

There were no serious acute or late radiation toxicities and this regmine was<br />

found as effective as the conventional arm.<br />

Arslan et al (12) designed a study to evaluate the effectiveness <strong>of</strong> tumor control<br />

with HRT in cases <strong>of</strong> GM and compare them with group previously treated<br />

with conventional radiotherapy concluded that no survival difference was seen<br />

between the two fractionation regimens. Survival in HRT arm was better but not<br />

statistically significant. Overall survival and progression free survival were 13.5<br />

and 11.3 months for HRT arm while it was 6.8 and 6.5 months for conventional<br />

arm.<br />

Phillips et al (20) had performed the first randomized comparison <strong>of</strong> conventional<br />

fractionated radiotherapy with short course <strong>of</strong> hyp<strong>of</strong>rectionated treatment in GM<br />

and AA A statistically significant survival difference between the two arms could<br />

not be demonstracted and they recommented further studies to support this result.<br />

Mc Aleese, et al (17) had treated two groups <strong>of</strong> poor prognosis malignant glioma<br />

patients one group with HRT 5 Gy/fraction biweekly up to 30 Gy the other group<br />

was treated with conventional RTH they concluded that HRT <strong>of</strong>fered reasonable,<br />

but lesser survival benefit than that would obtained with conventional RTH.<br />

In the randomized trial conducted at sites in seven European countries,<br />

researchers recruited 342 patients with newly diagnosed glioblastoma.<br />

Patients were assigned standard 60 Gy radiotherapy delivered in 2 Gy fractions<br />

for 6 weeks (n=100), hyp<strong>of</strong>ractionated 34 Gy radiotherapy delivered in 3,4 Gy<br />

fractions for 2 weeks (n=123), or 200 mg/m2 temozolomide daily on days 1 to<br />

5 every 4 weeks for 6 cycles (n=119).<br />

Median survival was 8.3 months for temozolomide, 7.5 months for<br />

hyp<strong>of</strong>ractionated radiation 34 Gy and 6 months for standard radiation. There<br />

was no significant difference in survival for patients treated with temozolomide<br />

or 34 Gy radiation.<br />

“These results indicate that standard 60 Gy radiation could replaced by HRT or<br />

temozolomide in patients with glioblastoma(21)<br />

In our study, we compare HRT with conventional radiotherapy in treatment <strong>of</strong><br />

GM patients. No survival difference was seen between both group but it was<br />

noticed that survival was slightly better in HRT group and <strong>of</strong> no statistical<br />

significance. Overall survival and progression free survival were 6.5 and 6<br />

months ,and 6 and 5 months for HRT and conventional radiotherapy groups<br />

respectively, and these results greatly coincide with the results <strong>of</strong> most trials<br />

which were mentioned in the literature.<br />

We concluded that HRT could be an option in treatment <strong>of</strong> GM patients where<br />

most <strong>of</strong> such patients are with poor performance status and the lesser number<br />

<strong>of</strong> fraction with HRT could be more suitable for them. Mostly HRT could be<br />

received three times a week or five times a week in the above mentioned studies<br />

a further studies is recommended to compare between both and choose the most<br />

effective. So studies with greater number <strong>of</strong> patients are advised to evaluate if<br />

HRT is an effective alternative treatment in GM and to determine the optimal<br />

fraction size when HRT is used as an adjuvant treatment <strong>of</strong> GM.<br />

References<br />

1. Saleman M. Glioblastoma multiforme and anaplatic astrocytoma. In: Kaye<br />

AH, Laws ER (eds) : Brain Tumors : An encyclopedic approach Lnhurchill<br />

Livingstone, 2001, PP 493 – 523 .<br />

2. Schoenberg BS, Schoenberg DC, Christine BW. The epidemiology <strong>of</strong><br />

primary intracranial neoplasms <strong>of</strong> childhood : A population study. Mayo<br />

Clin proc 1976; 51 : 51 -56 .<br />

3. Chadha M, Cpala J, Cderre JA, et al. Boron – neutron – capture therapy for<br />

glioblastoma multiforme using the epithermal neutron beam at Brook haven<br />

National laboratory. Int J Radiat oncol Biol Phys 1998; 40 : 929 – 834 .<br />

4. Bakhemuka M; Massey E; Schoenberg B International mortality from<br />

primary nervous system neoplasms distribution and trends. Int, J Epidem<br />

1988; 17 : 33 – 38 .<br />

5. Lederman G; Arbit E, Odaimi M, et al., Fractionated stereotactic<br />

radiosurgery and concurrent taxol in recurrent glioblastoma multiforme : A<br />

preliminary report. Int. J. Radiat. Oncol Biol Phys 1998; 40 (3) : 661 – 666 .<br />

6. Walker MD. Green SB, Byar DP, et al Randomized comparisons <strong>of</strong><br />

radiotherapy and nitrosoureas for the treatment <strong>of</strong> malignant glioma after<br />

surgery : BTSG 22 – 01. N Engl J Med 1980; 303 : 1323 – 1327 .<br />

7. Nakagawa K, Aoki Y, Fujimaki T, et al. High-dose conformal radiotherapy<br />

influenced the pattern <strong>of</strong> failure but did not improve survival in glioblastoma<br />

multiforme. Int J Radiat Oncol Biol Phys 1998; 40 : 1141 – 1149 .<br />

8. Nelson DF, Cuman WJ, Scott C, et al. Hyperfractionated radiation therapy<br />

and bis-chloroethyl nitrosourea in the treatment <strong>of</strong> malignant glioma –<br />

Possible advantage observed at 72 Gy on 1.2 BID fractions : Report <strong>of</strong> the<br />

RTOG protocol 8302 . Int J Radiat Oncol Biol Phys 1993; 25 : 193 – 207 .<br />

50 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


9. Nathan SF, Shiao YW, Bin ST, et al. Hyp<strong>of</strong>ractionated intensity – modulated<br />

radiotherapy for primary glioblastoma multiforme. No 3, PP 721 – 726 .<br />

10. Glinski B. Postoperative hyp<strong>of</strong>ractionated radiotherapy versus<br />

conventionally fractionated radiotherapy in malignant gliomas : A<br />

preliminary report on a randomized trial J Neuro Oncol 1993; 16 : 167 –<br />

172 .<br />

11. Shepherd SF, Laing RW, Cosgrove VP, etal. Hyp<strong>of</strong>ractionated stercotactic<br />

radiotherapy in the management <strong>of</strong> recurrent glioma. Int J Radiat Oncol Biol<br />

Phys 1997; 37 : 393 – 398 .<br />

12. Arslan AN, Karadeniz G, Guveli MF. Postoperative hyp<strong>of</strong>ractionated<br />

radiotherapy in glioblastoma multiforme. <strong>Journal</strong> <strong>of</strong> BUON 2006; 11 : 39<br />

– 42 .<br />

13. Slotman BJ, Kralendonk JH, Van Alphen HA, et al. Hyp<strong>of</strong>ractionated<br />

radiation therapy in patents with glioblastoma multiforme : Results <strong>of</strong><br />

treatment and impact <strong>of</strong> prognostic factors. Int J Radiat Oncol Biol Phys<br />

1996; 34 : 895 – 898 .<br />

14. Laperrieve N, Zuraw L, Cairncross G et al. Radiotherapy for newly<br />

diagnosed malignant glioma in adults : A systematic review. Radiother.<br />

Oncol 2002; 64 : 259 – 273 .<br />

15. MRC Brain Tumor Working Party, A medical research council trial<br />

astrocytoma. Br J Cancer 1991; 64 (4) : 769 – 774 .<br />

16. Chan JL, Lee SW, Fraass BA et al. Survival and failure <strong>of</strong> high grade<br />

gliomas after three dimensional conformal radiotherapy. J Clin Oncol 2002;<br />

20 : 1635 – 42 .<br />

17. McALeese JJ, Stenning SP, Ashley S et al. Hyp<strong>of</strong>ractionated radiotherapy<br />

for poor prognosis malignant glioma : matched pair survival analysis with<br />

MRC controls. Radiotherapy α <strong>Oncology</strong> 67 (2003) 177 – 182.<br />

18. Slotman B J, Kralendonk JH, van Alphen HAM et al. Hyp<strong>of</strong>ractionated<br />

radiation therapy in patients with glioblastome multiform : Results <strong>of</strong><br />

treatment and impact <strong>of</strong> prognostic factors. Int J Radiat Oncal Bid Phys<br />

1996; 34 : 895 – 898 .<br />

19. Lang O, liebermeister E, Liesegang J, et al. Radiotherapy <strong>of</strong> glioblastoma<br />

multiforme : Feasibility <strong>of</strong> increased fraction size and shortened overall<br />

treatment. Strahlenther Onkol 1998; 174 : 629 – 632 .<br />

20. Phillips C, Guiney M, Smith J, et al. A randomized trial comparing 35 Gy in<br />

ten fractions <strong>of</strong> cerebral irradiation for glioblastoma multiforme and older<br />

patients with anaplastic astrocytoma. Radio therapy α oncology 68 (2003)<br />

23 – 26 .<br />

21. A Malmstrom, B. H. Grønberg, R. Stupp, C. Marosi, D. Frappaz et al<br />

glioblastoma (GBM) in elderly patients: A randomized phase III trial<br />

comparing survival in patients treated with 6-week radiotherapy (RT)<br />

versus hyp<strong>of</strong>ractionated RT over 2 weeks versus temozolomide single-agent<br />

chemotherapy (TMZ) J Clin Oncol 28:18s, 2010 (suppl; abstr LBA2002)<br />

Characteristics<br />

Age<br />

50<br />

Sex<br />

male<br />

female<br />

ECOG<br />

Duration <strong>of</strong><br />

symptoms<br />

10week<br />

Increase ICT<br />

Present<br />

absent<br />

Surgery<br />

Complete<br />

Subtotal<br />

Biopsy<br />

Table 1 shows patient characteristics<br />

Hyp<strong>of</strong>ractionated<br />

Conventional<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 51<br />

NO<br />

9<br />

11<br />

12<br />

8<br />

7<br />

13<br />

14<br />

6<br />

16<br />

4<br />

12<br />

5<br />

3<br />

Table 2: Difference between the study group and control groups were non<br />

significant statistically<br />

Survival<br />

Median – over all survival<br />

Progression free survival<br />

%<br />

45<br />

55<br />

60<br />

40<br />

35<br />

65<br />

70<br />

30<br />

80<br />

20<br />

60<br />

25<br />

15<br />

Hyp<strong>of</strong>ractionated<br />

6.5 month<br />

6 month<br />

NO<br />

6<br />

14<br />

13<br />

7<br />

11<br />

9<br />

12<br />

8<br />

17<br />

3<br />

10<br />

6<br />

4<br />

Conventional<br />

6 month<br />

5 month<br />

%<br />

30<br />

70<br />

65<br />

35<br />

55<br />

45<br />

60<br />

40<br />

85<br />

15<br />

50<br />

30<br />

20<br />

P – value<br />

0.509<br />

0.967


original article <<br />

Percent survival<br />

Fig 1: over all survival<br />

Percent survival<br />

100<br />

75<br />

50<br />

25<br />

100<br />

Fig 2: Progression free survival<br />

Overall Survival<br />

0<br />

0 5 10<br />

Time (months)<br />

15 20<br />

75<br />

50<br />

25<br />

Progression free survival<br />

0<br />

0 6 12<br />

Time<br />

Hyp<strong>of</strong>ractiontion<br />

Conventional<br />

Hyp<strong>of</strong>ractiontion<br />

Conventional<br />

52 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org<br />

N


New<br />

vinflunine<br />

The 1 st and only registered chemotherapy<br />

after failure <strong>of</strong> a platinum-containing regimen<br />

in advanced or metastatic TCCU<br />

www.amaac.org Pierre Fabre <strong>Oncology</strong> Middle East - Riad El Solh - P.O.Box 11 - 2131 Beirut - Lebanon <strong>Pan</strong> <strong>Arab</strong> Fax : <strong>Journal</strong> 00 961 <strong>of</strong> 1 98 <strong>Oncology</strong> 98 42 | vol 4; issue 1 | March 2011 < 53<br />

Full Prescribing information is available upon request<br />

Æ


news from the arab world <<br />

Announcements<br />

Dear all,<br />

We would like to announce the creation <strong>of</strong> the<br />

Junior Oncologists (JAMAAC)<br />

gathering all over the <strong>Arab</strong> world under the umbrella <strong>of</strong> <strong>Arab</strong><br />

<strong>Medical</strong> <strong>Association</strong> <strong>Against</strong> Cancer and includes every young<br />

doctor working against cancer (medical, surgical,radiation,<br />

basic sciences, etc...)<br />

We have established this group to serve YOU and invest<br />

in YOU. Building capacities and facilitating mobility and<br />

research grants to all <strong>of</strong> YOU either through AMAAC<br />

fellowships or other internationally available grants that we<br />

will keep on announcing. Also, we will organize a number <strong>of</strong><br />

educational and scientific activities to be announced soon. We<br />

look forward for your active participation.<br />

This group is totally endorsed by AMAAC board and<br />

Secretary General Pr<strong>of</strong>. Sami Khatib.<br />

So i would like all <strong>of</strong> you who have facebook accounts to<br />

add Dr. Sami Khatib with the user name <strong>of</strong> Sami Khatib on<br />

Facebook for all the social activities.<br />

And to join the group <strong>of</strong> JAMAAC on Facebook with the<br />

user name <strong>of</strong> : ناطرسلا ةحفاكلم برعلا ءابطلأا بابش ةطبار - Junior<br />

AMAAC (JAMAAC).<br />

Waiting to see all <strong>of</strong> you members <strong>of</strong> this important group.<br />

Thanks, Majd Alkhatib<br />

Dear colleagues,<br />

The Abstract Submission Deadline for the 8th International AORTIC conference<br />

is upon us and is scheduled to close on the 15th May 2011. For those still busy<br />

with research, Late Breaking Abstracts will be accepted between 1 – 15 October<br />

2011.<br />

As indicated in our previous communiqué, we have been monitoring the<br />

situation in Egypt closely and taking everything into consideration we would<br />

like to reiterate that the conference will still be going ahead in Cairo , Egypt ,<br />

as scheduled.<br />

We are also pleased to inform you that the development <strong>of</strong> Program is going<br />

well and is at an advanced stage, to date over 300 Abstracts have been received.<br />

Therefore the content and program promises to be highly interesting and<br />

informative.The conference promises to be excellent on all counts, if you have<br />

not registered yet, we urge you to register now.<br />

Please do not hesitate to contact us should you require any further information.<br />

Kind Regards<br />

AORTIC Conference Secretariat<br />

Global Conferences Africa - P O Box 6761, Roggebaai, Cape Town, South Africa<br />

Tel.: +27 21 408 9988<br />

Fax: +27 21 408 9956<br />

E-mail: aortic@globalconf.co.za<br />

54 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


Under the high patronage <strong>of</strong> his excellence<br />

Zine El Abidine Ben Ali, president <strong>of</strong> the republic <strong>of</strong> Tunisia,<br />

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

organizes in Tunisia, in Hammamet (Hotels Russelior and Royal)<br />

from April 29th to May 1st 2011,<br />

the 11th <strong>Pan</strong>arab Cancer Congress<br />

The topic concern: breast, lung, urologic and gynecologic<br />

cancers, hematologic malignancies and palliative care within<br />

lectures done by <strong>Arab</strong> and international recognized experts in<br />

the field.<br />

Posters sessions are programmed.<br />

Abstracts dealdline is fixed to the end <strong>of</strong> february<br />

You may fastly have all the informations via the website<br />

www.amaactunisia.com<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 55


news from the arab world <<br />

56 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 57


news from the arab world <<br />

58 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


FLYER MCO_RV (7 Feb).qxp 21-04-2011 15:46 Pagina 3<br />

EURO-ARAB SCHOOL OF ONCOLOGY MASTERCLASS<br />

3 R D E A S O M A ST E RCLASS IN<br />

C L I N I CAL ONCOLO GY<br />

Jointly organised with the<br />

<strong>Arab</strong> <strong>Medical</strong> <strong>Association</strong><br />

Ag a i n st Cancer (AMAAC)<br />

27-29 October 2011<br />

Amman, J o rd a n<br />

Chair: N. Pavlidis, GR - H. Khaled, EG<br />

Scientific coordinators: S. Khatib, JO - A. Costa, IT<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 59


news from the arab world <<br />

60 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


3rd ASIAN<br />

Breast Cancer Congress<br />

Interdisciplinary Integration<br />

3 - 4 MARCH, 2012 / BANGALORE, INDIA<br />

Supported By<br />

adding life to years<br />

www.amaac.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 61


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 />

62 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


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<br />

region 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<br />

description <strong>of</strong> the study design, conduct, and analysis methods<br />

used to obtain 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.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 63


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 e-mail at the following e-mail:<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 />

64 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


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<br />

nevi as precursor lesions and risk markers <strong>of</strong> melanoma, in<br />

Kirkwood JM (ed): Molecular Diagnosis and Treatment <strong>of</strong><br />

Melanoma. New 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<br />

years <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<br />

<strong>of</strong> two multicenter, randomized, placebo-controlled studies <strong>of</strong><br />

pamidronate disodium for the palliation <strong>of</strong> bone pain in men with<br />

metastatic 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<br />

for each contributing author at manuscript submission. Upon<br />

acceptance <strong>of</strong> the manuscript, each author will receive an e-mail<br />

invitation to sign a statement confirming that the manuscript<br />

contains no material for which publication would violate any<br />

copyright or other personal or proprietary right <strong>of</strong> any person or<br />

entity. Manuscripts will not be published until each author has<br />

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.org <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 < 65


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 E-mail 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 />

66 > <strong>Pan</strong> <strong>Arab</strong> <strong>Journal</strong> <strong>of</strong> <strong>Oncology</strong> | vol 4; issue 1 | March 2011 www.amaac.org


e<br />

ns<br />

ria<br />

ns<br />

led<br />

ith<br />

.g.,<br />

ble<br />

ery<br />

in,<br />

ing<br />

ell<br />

sia<br />

RF<br />

ed<br />

tic<br />

gin<br />

en<br />

ally<br />

not<br />

n a<br />

ory<br />

om<br />

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

on.<br />

en:<br />

: 1<br />

2;<br />

ge:<br />

: 4<br />

0;<br />

er:<br />

is<br />

ug,<br />

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

p ®<br />

x ®<br />

3.<br />

16:16


GLI Ad-1 Mar 10 JOR

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!