Designed at National Institute of Design, Ahmedabad.
Publications > Scientific Papers
Scientific Papers Books Manuals




Women's perceptions and social barriers determine compliance to cervical screening: Results from a population based study in India

Partha Basu MDa, Srabanti Sarkar MAa, Supriya Mukherjee MSWa, Mousumi Ghoshal MSWa, Srabani Mittal MBBS DPHa, Sutapa Biswas BAb, Ranajit Mandal MDa, Rengaswamy  Sankaranarayanan MDc

aDepartment of Gynecologic Oncology, Chittaranjan National Cancer Institute, 37 S. P. Mukherjee Road, Kolkata 700026, India,
b Cancer Foundation of India, Kolkata, India,
c Screening Group, International Agency for Research on Cancer, Lyon, France,

Cancer Detection and Prevention , 30 (4), 369-74, 2006

Abstract:

Background:


Success of cervical screening initiatives depends on high participation of the target population, which in turn is determined by the women's perceptions, health orientation and other socio-cultural issues. The present study identifies the immediate social and cultural barriers that prevent women to attend cervical screening facilities.

Methods:

Women non-compliant to a community-based cervical screening program were identified. From them 500 were randomly selected for interview using a structured questionnaire that was designed on the basis of feedbacks received from several focused group discussions. Questionnaire listed 24 possible reasons for non-compliance. The women were asked to select the most pertinent reason(s) for her non-attendance or to reveal if they had any reason other than the listed ones.

Results:

A total of 469 non-compliant women were interviewed. They had significantly lower literacy rate compared to the compliant women (OR = 2.25; 95% CI: 1.23-4.13). Nearly half of the interviewed women responded that they themselves opted to stay away from the program. Most common reasons cited for non-attendance in this group were reluctance to go for medical test in the absence of any symptoms and apprehension to have a test that detects cancer. Second major group of responders comprised of women who were willing, yet could not attend due to various hurdles. Most common hurdles were inability to leave household chores, pre-occupation with family problems and lack of approval from husbands.

Conclusions:

Modification of health behavior through education and social empowerment of women are essential for a population based cervical screening program to succeed in India.


Molecular variants of HPV-16 associated with cervical cancer in Indian population

M.R. Pillai1, R. Hariharan1, Janki Mohan Babu1, S. Lakshmi2, S.V. Chiplunkar3,4, M. Patkar3,4, H. Tongaonkar3,4, K. Dinshaw3,4, R.S. Jayshree5, B.K.M. Reddy5, M. Siddiqi6, Soma Roychowdhury6, Baisakhi Saha7, P. Abraham8, M. Gnanamony8, A. Peedicayil9, J. Subhashini10, T.S. Ram10, Bindu Dey11, C. Sharma12, S.K. Jain13, N. Singh12,7

1Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India,
2Regional Cancer Centre, Thiruvananthapuram, India,
3Tata Memorial Center, Parel, Mumbai, India,
4Advanced Centre for Treatment, Research, Education in Cancer, Kharghar, Navi Mumbai, India,
5Department of Microbiology, Kidwai Memorial Institute of Oncology, Bangalore, India,
6Cancer Foundation of India, Kolkata, India,
7Bose Institute, Kolkata, India,
8Department of Clinical Virology, Christian Medical College, Vellore, India,
9Department of Obstetrics and Gynaecology, Christian Medical College, Vellore, India,
10Department of Radiation Oncology, Christian Medical College, Vellore, India,
11Department of Biotechnology, New Delhi, India,
12Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India,
13Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, India,

Abstract:

Human papilloma virus is a causative factor in the etiology of cervical cancer with HPV16 being the most prevalent genotype associated with it. Intratype variations in oncogenic E6/E7 and capsid L1 proteins of HPV 16 besides being of phylogenetic importance, are associated with risk of viral persistence and progression. The objective of this multicentric study was to identify HPV-16 E6, E7 and L1 variants prevalent in India and their possible biological effects. Squamous cell cervical cancer biopsies were collected from 6 centres in India and examined for the presence of HPV 16. Variants of HPV-16 were characterized by full length sequence analysis of L1, E6 and E7 genes in 412 samples. Similar distribution of the variants was seen from the different centres/regions, with the European variant E350G being the most prevalent (58%), followed by American Asian variant (11.4%). Fifty six changes were seen in E6 region, 31 being nonsynonymous. The most frequent being L83V (72.3%), Q14H (13.1%) and H78Y (12.1%). Twenty-nine alterations were seen in E7 region, with 12 being nonsynonymous. The most frequent being F57V (9%). L1 region showed 204 changes, of which 67 were nonsynonymous. The most frequent being 448insS (100%), and 465delD (100%), H228D (94%), T292A (85%). The identified variants some new and some already reported can disrupt pentamer formation, transcriptional regulation of the virus, L1 protein interface interaction, B and T cell epitopes, p53 degradation, and thus their distribution is important for development of HPV diagnostics, vaccine, and for therapeutic purpose. © 2009 UICC


Human Papillomavirus Genotype Distribution in Cervical Cancer in India: Results from a Multi-center Study

Partha Basu1, Soma Roychowdhury2, Uttam Das Bafna3, Santanu Chaudhury4, Sarita Kothari4, Rupinder Sekhon5, Dhananjaya Saranath6, Sutapa Biswas2, Petter Gronn7, Ivan Silva7, Maqsood Siddiqi2, Sam Ratnam8

1Department of Gynecologic Oncology, Chittaranjan National Cancer Institute, Kolkata,
2Cancer Foundation of India, Kolkata,
3Kidwai Memorial Institute of Cancer, Bangalore,
4RST Regional Cancer Center, Nagpur,
5Rajiv Gandhi Institute of Cancer, New Delhi,
6Reliance Life Sciences, Mumbai, India,
7Norchip, Klokkarstua, Norway,
8Public Health Laboratory, St. John’s, Canada,

Asian Pacific J Cancer Prev, 10, 27-34, 2009

Abstract:

The prevalence of HPV genotypes in cervical cancer differs in various regions, though types 16 and 18 generally account for the majority. Knowledge of HPV genotypes in cervical cancer covering the diverse Indian population is important in consideration of the potential future impact of HPV prophylactic vaccination and HPV-based screening strategies. To determine HPV genotype distribution in cervical cancers representing different regions a total of 278 cervical cancer cases were enrolled from cancer centers in North, East, Central and South India. Cervical scrape specimens were tested for HPV DNA using the MY09/11 L1 consensus PCR method followed by sequencing for enotyping, as well as for HPV mRNA utilizing the PreTectTM HPV-Proofer assay. In instances of negative or discrepant results between the two tests, biopsy specimens were tested. HPV DNA and/or mRNA were detected in 91.7% of the cases. Genotype 16 was the most common type, detected alone in 59.4% and in association with type 18 in 3.6% of cases. Genotype 18 was detected as a monotype in 13.3% cases. In total, types 16 and 18 alone or in co-infection with each other were detected in 76.3% cases. Genotype 33 was the third most common type. Overall, genotypes 16, 18, 31, 33, and 45 were the five most common types, detected in 87.1% of the total cases. There were no significant regional differences. In conclusion, the currently available HPV prophylactic vaccines targeting types 16 and 18 have the potential to reduce the burden of cervical cancer in India by over 75%.


Region-Wise Distribution of High-Risk Human Papillomavirus Types in Squamous Cell Carcinomas of the Cervix in India

Pillai MR1, Babu JM1, Jissa VT1, Lakshmi S2, Chiplunkar SV3,4, Patkar M3,4, Tongaonkar H3,4, Reddy BKM5, Kumar Chakka6, Siddiqi M7, Roychowdury S7, Abraham P8, Peedicayil A9, Gnanamony M8, Subashini J10, Ram TS10, Bindu Dey11, Singh N12, Singh A12, Jain SK13 and Jayshree RS6,14

1Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India,
2Regional Cancer Centre, Thiruvananthapuram, India,
3Tata Memorial Center, Parel, Mumbai, India,
4Advanced Centre for Treatment, Research, Education in Cancer, Kharghar, Navi Mumbai, India,
5Department of Radiotherapy, Kidwai Memorial Institute of Oncology, Bangalore, India,
6Department of Microbiology, Kidwai Memorial Institute of Oncology, Bangalore, India,
7Cancer Foundation of India, Kolkata, India,

8Department of Clinical Virology, Christian Medical College, Vellore, India,
9Department of Obstetrics and Gynaecology, Christian Medical College, Vellore, India,
10Department of Radiation Oncology, Christian Medical College, Vellore, India,
11Department of Biotechnology, New Delhi, India,
12Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India,
13Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, India,

International Journal of Gynecological Cancer. 20(6):1046-1051, August 2010.


Abstract:

Hypothesis:

Assessment of the prevalence and type distribution of human papillomavirus (HPV) in squamous cell carcinomas (SCC) of the cervix across India was undertaken to estimate the impact of available prophylactic HPV-L1 vaccines in the country and to find out additional types that might be needed to be incorporated in second-generation vaccines.

Methods:

High-risk (HR) HPVs were genotyped from 667 histopathologically confirmed cases of SCC from 6 different centers representing 4 regions across India: Advanced Centre for Treatment, Research and Education in Cancer, Mumbai; All India Institute of Medical Sciences, New Delhi; Cancer Foundation of India, Kolkata; Christian Medical College, Vellore; Kidwai Memorial Institute of Oncology, Bangalore; and Regional Cancer Center, Thiruvananthapuram. Human papillomaviruses in tumor biopsies were analyzed by Xcytonscreen HPV based on PGMY09/11 multiplex polymerase chain reaction and reverse dot blot assay.

Results:

Overall viral prevalence across India was not different; 92.1% of 667 cases harbored HPV; 8% were negative. Infection with single HR type was seen in 86.8%: predominant types being HPV-16 followed by HPV-18, -45, -73, -31, -56, -52, -58, -59, -33, -68, -51, -35, -26, and -39. Human papillomavirus types 16/18-positive fraction formed 79.6%; other types comprised 12.4%.

Conclusions:

Prophylactic HPV-16/18-L1 vaccines would provide greater than 75% protection against SCC in India. Ranking and frequencies of non-16/18 types were different from earlier reports. Hence, considering the possibility of promotion of persistence of nonvaccine types in the vaccinees due to original antigenic sin and the lack of organized screening programs in India, a broad-based vaccine approach would be appropriate.


Curcumin protects DNA damage in a chronically arsenic-exposed population of West Bengal

Jaydip Biswas1, Dona Sinha2, Sutapa Mukherjee2, Soumi Roy2, Maqsood Siddiqi3, Madhumita Roy2

Human Experimental Toxicology, 29: 513-524


1Chittaranjan National Cancer Institute, Kolkata, Department of Environmental,
2Carcinogenesis and Toxicology, Chittaranjan National Cancer Institute, Kolkata, India,
3Cancer Foundation of India, Kolkata, India,

Abstract:

Groundwater arsenic contamination has been a health hazard for West Bengal, India. Oxidative stress to DNA is recognized as an underlying mechanism of arsenic carcinogenicity. A phytochemical, curcumin, from turmeric appears to be potent antioxidant and antimutagenic agent. DNA damage prevention with curcumin could be an effective strategy to combat arsenic toxicity. This field trial in Chakdah block of West Bengal evaluated the role of curcumin against the genotoxic effects of arsenic. DNA damage in human lymphocytes was assessed by comet assay and fluorescence-activated DNA unwinding assay. Curcumin was analyzed in blood by high performance liquid chromatography (HPLC). Arsenic induced oxidative stress and elucidation of the antagonistic role of curcumin was done by observation on reactive oxygen species (ROS) generation, lipid peroxidation and protein carbonyl. Antioxidant enzymes like catalase, superoxide dismutase, glutathione reductase, glutathioneS-transferase, glutathione peroxidase and non-enzymatic glutathione were also analyzed. The blood samples of the endemic regions showed severe DNA damage with increased levels of ROS and lipid peroxidation. The antioxidants were found with depleted activity. Three months curcumin intervention reduced the DNA damage, retarded ROS generation and lipid peroxidation and raised the level of antioxidant activity. Thus curcumin may have some protective role against the DNA damage caused by arsenic.


MassARRAY Spectrometry Is More Sensitive than PreTect HPV-Proofer and Consensus PCR for Type-Specific Detection of High-Risk Oncogenic Human Papillomavirus Genotypes in Cervical Cancer

Basu P1, Chandna P2, Bamezai RN3, Siddiqi M4, Saranath D5, Lear A6, Ratnam S7

1Chittaranjan National Cancer Institute, Kolkata, India,
2AceProbe Technologies (India) Pvt. Ltd., New Delhi, India,
3School of Life Sciences, Jawaharlal Nehru University, New Delhi, India,
4Cancer Foundation of India, Kolkata, India,
5Reliance Life Sciences, Mumbai, India,
6Dr. H. Bliss Murphy Cancer Centre, St. John's, Newfoundland and Labrador, Canada,
7Public Health Laboratory, St. John's, Newfoundland and Labrador, Canada,

J Clinical Microbiology, 49, 3537-44, 2011

Abstract
:


Type-specific detection of human papillomavirus (HPV) is indicated for better risk stratification and clinical management of women testing positive for HPV and for epidemiologic surveillance. MassARRAY spectrometry (MassARRAY; Sequenom) is a novel method for type-specific detection of 15 high-risk oncogenic HPV types: HPV-16, -18, -31, -33, -35, -39, -45, -51, -52, -56, -58, -59, -66, -68, and -73. PreTect HPV-Proofer (Proofer; Norchip) is a type-specific assay that detects E6/E7 mRNA from five high-risk oncogenic HPV types: HPV-16, -18, -31, -33, and -45. The performance of these tests for type-specific identification of HPV was assessed with cervical specimens from 192 cases of cervical cancer in comparison with consensus MY09/MY11 PCR followed by nucleotide sequencing (consensus PCR). The overall HPV detection rates were 94.8% (95% confidence interval [CI], 91.7, 97.9), 83.3% (95% CI, 78.1, 88.5), and 86.5% (95% CI, 81.7, 91.3) for MassARRAY, Proofer, and consensus PCR, respectively. All tests were negative in six (3.1%) of the 192 cases. Considering only the specimens that contained at least one of the five types targeted by Proofer, the detection rates were 96.6%, 91.4%, and 86.9% for MassARRAY, Proofer, and consensus PCR, respectively. MassARRAY detected multiple infections in 14.1%, Proofer detected multiple infections in 3.6%, and consensus PCR failed to detect any multiple infections. The agreement was highest at 86.0% (kappa = 0.76) between MassARRAY and Proofer and lowest at 81.8% (kappa = 0.69) between Proofer and consensus PCR. In conclusion, MassARRAY is a highly sensitive and accurate method for type-specific detection of oncogenic HPV in cervical cancer, with Proofer showing impressive performance.


Pictorial warnings on Tobacco Products. (Research letter)

Mira Aghi1, Kunal Oswal2,3, Mangesh Pednekar3, Alexander Cyril 4, Stupa Biswas5 .

1Behavioral Scientist and a Communication freelancer, Delhi, New Delhi, India;
2Public Health Dentistry, Terna Dental College, Mumbai, Maharashtra, India;
3Healis-Sekhsaria Institute for Public Health, Mumbai,Maharashtra, India;
4Mary Anne Charity Trust, Chennai,Tamil Nadu, India;
5Cancer Foundation of India, Kolkatta.

BMJ Tobacco Control, 10.1136/tobaccocontrol-2011-050035.

Abstract:

According to the 2009 Government of India notification, all tobacco products should carry pictorial messages1 and a revised statutory warning ‘Smoking kills and tobacco causes cancer’, on at least 40%2 of the principal display areas. For non-compliance, the dealer or seller can be fined up to Rs 1000 (wUS$20) with or without a 1-year imprisonment. If the offence is repeated, the fine amount will be raised to Rs 3000 (US$60) with or without a 2-year imprisonment.3 There was no explicit mention in the law that warnings were to be visible at the point of sale. Although the law requires that the warnings cannot be severed or covered when the package is sealed or opened, there was no explicit requirement that the warnings be made visible at the point of sale. The aim of this study was to examine the visibility of tobacco products’ pictorial health warnings at the point of sale.


Proceedings of the Indo-U.S. bilateral workshop on accelerating botanicals/biologics agent development research for cancer chemoprevention, treatment, and survival. (Meeting report)

Nagi B. Kumar1, Medha Dhurandhar2, Bharat Aggarwal3, Shrikant Anant4, Kenyon Daniel1, Gary Deng5, Julie Djeu1, Jinhui Dou6, Ernest Hawk3, B. Jayaram7, Libin Jia8, Rajendra Joshi9, MadhuriKararala10, Devarajan Karunagaran11, Omer Kucuk12, Lalit Kumar13, Mokenge Malafa1, G. J.Samathanam14, Fazlul Sarkar15, Maqsood Siddiqi16, Rana P. Singh17, Anil Srivastava18 & Jeffrey D. White8

1 Moffitt Cancer Center, Tampa, Florida, 33612-9497;
2 Centre for Development of Advanced Computing, Pune University, Pune, 411007, India;
3 The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, 77054;
4 The University of Kansas Medical Center, Kansas City, Kansas, 66160;
5 Memorial Sloan-Kettering Cancer Center, New York, New York, 10021;
6 Food and Drug Administration, Silver Springs, Maryland, 20993;
7 India Institute of Technology-Delhi, New Delhi, 110016, India;
8 National Cancer Institute, NIH, Bethesda, Maryland, 20892;
9 Bioinformatics Scientific and Engineering Computing, Pune University, Pune, 411007, India;
10 University of Michigan, Ann Arbor, Michigan, 48109-5930;
11 Department of Biotechnology, India Institute of Technology – Madras, Chennai, 600036, India;
12 Emory Healthcare, The Emory Clinic Winship Cancer Institute, NE Atlanta, Georgia, 30322;
13 Institute Rotary Cancer Hospital (IRCH), All India Institute of Medical Sciences, New Delhi, 110029, India;
14 Department and Transfer Division, Department of Science and Technology, Government of India, India;
15 Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, 48201;
16 Cancer Foundation of India, Tiljala, Kolkata, 700039, India;
17 School of Life Sciences, Central University of Gujarat, Gujarat, 382030, India;
18 Open Health Systems Laboratory at Johns Hopkins Montgomery County Campus, Rockville, Maryland, 20850

Cancer Medicine 2013; 2(1): 108–115

Abstract:

With the evolving evidence of the promise of botanicals/biologics for cancer chemoprevention and treatment, an Indo-U.S. collaborative Workshop focusing on “Accelerating Botanicals Agent Development Research for Cancer Chemoprevention and Treatment” was conducted at the Moffitt Cancer Center, 29–31 May 2012. Funded by the Indo-U.S. Science and Technology Forum, a joint initiative of Governments of India and the United States of America and the Moffitt Cancer Center, the overall goals of this workshop were to enhance the knowledge (agents, molecular targets, biomarkers, approaches, target populations, regulatory standards, priorities, resources) of a multinational, multidisciplinary team of researcher’s to systematically accelerate the design, to conduct a successful clinical trials to evaluate botanicals/biologics for cancer chemoprevention and treatment, and to achieve efficient translation of these discoveries into the standards for clinical practice that will ultimately impact cancer morbidity and mortality. Expert panelists were drawn from a diverse group of stakeholders, representing the leadership from the National Cancer Institute’s Office of Cancer Complementary and Alternative Medicine (OCCAM), NCI Experimental Therapeutics (NExT), Food and Drug Administration, national scientific leadership from India, and a distinguished group of population, basic and clinical scientists from the two countries, including leaders in bioinformatics, social sciences, and biostatisticians. At the end of the workshop, we established four Indo-U.S. working research collaborative teams focused on identifying and prioritizing agents targeting four cancers that are of priority to both countries. Presented are some of the key proceedings and future goals discussed in the proceedings of this workshop.


European guidelines for quality assurance in colorectal cancer screening and diagnosis: Overview and introduction to the full Supplement publication. (Guideline)

European Colorectal Cancer Screening Guidelines Working Group:

L. von Karsa1, J. Patnick2,3, N. Segnan1,4,W. Atkin5, S. Halloran6,7, I. Lansdorp-Vogelaar8, N. Malila9, S. Minozzi4, S. Moss10, P. Quirke11, R. J. Steele12, M. Vieth13, L. Aabakken14, L. Altenhofen15, R. Ancelle-Park16, N. Antoljak17,18, A. Anttila9, P. Armaroli4, S. Arrossi19, J. Austoker20, †, R. Banzi21, C. Bellisario4, J. Blom22, H. Brenner23, M. Bretthauer24, M. Camargo Cancela25,26, G. Costamagna27, J. Cuzick28, M. Dai29, J. Daniel26,30, E. Dekker31, N. Delicata32, S. Ducarroz1, H. Erfkamp33, J. A. Espinàs34, J. Faivre35, L. Faulds Wood36, A. Flugelman37, S. Frkovic-Grazio38, B. Geller39, L. Giordano4, G. Grazzini40, J. Green20, C. Hamashima41, C. Herrmann26,42, P. Hewitson20, G. Hoff43,44, I. Holten45, R. Jover46, M. F. Kaminski47, E. J. Kuipers8, J. Kurtinaitis48, †, R. Lambert1, G. Launoy49,W. Lee50, R. Leicester51, M. Leja52, D. Lieberman53, T. Lignini1, E. Lucas1, E. Lynge54, S. Mádai55, J. Marinho56, J. Maučec Zakotnik57, G. Minoli58, C. Monk59, A. Morais60, R. Muwonge1, M. Nadel61, L. Neamtiu62, M. Peris Tuser63, M. Pignone64, C. Pox65, M. Primic-Zakelj66, J. Psaila32, L. Rabeneck67, D. Ransohoff64, M. Rasmussen68, J. Regula47, J. Ren26, G. Rennert37, J. Rey69, R. H. Riddell70, M. Risio71, V. Rodrigues72, H. Saito41, C. Sauvaget1, A. Scharpantgen73,W. Schmiegel65, C. Senore4, M. Siddiqi74, D. Sighoko26,75, R. Smith30, S. Smith76, S. Suchanek77, E. Suonio1, W. Tong78, S. Törnberg79, E. Van Cutsem80, L. Vignatelli81, P. Villain20, L. Voti26,82, H.Watanabe83, J.Watson20, S.Winawer84, G. Young85, V. Zaksas86, M. Zappa40, R. Valori87

1 International Agency for Research on Cancer, Lyon, France;
2 NHS Cancer Screening Programmes Sheffield, United Kingdom;
3 Oxford University Cancer Screening Research Unit, Cancer Epidemiology Unit,University of Oxford, Oxford, United Kingdom;
4 CPO Piemonte, AO Città della Salute e della Scienza di Torino, Turin Italy;
5 Imperial College London, London, United Kingdom;
6 Bowel Cancer Screening Southern Programme Hub, Royal Surrey County Hospital NHS Foundation Trust, Guildford, United Kingdom;
7 University of Surrey, Guildford, United Kingdom;
8 Erasmus MC, Rotterdam, the Netherlands;
9 Finnish Cancer Registry, Helsinki, Finland;
10 The Institute of Cancer Research, Royal Cancer Hospital, Sutton, United Kingdom;
11 Leeds Institute of Molecular Medicine, St James’ University Hospital, Leeds, United Kingdom;
12 Ninewells Hospital and Medical School, Dundee, United Kingdom;
13 Institute of Pathology, Klinikum Bayreuth, Bayreuth, Germany;
14 Department of Medical Gastroenterology, Stavanger University Hospital, Stavanger, Norway;
15 Central Research Institute of Ambulatory Health Care, Berlin, Germany;
16 Direction Générale de la Santé, Paris, France;
17 Croatian National Institute of Public Health, Zagreb, Croatia;
18 University of Zagreb School of Medicine, Zagreb, Croatia;
19 CONICET/CEDES, Buenos Aires, Argentina;
20 University of Oxford, Oxford, United Kingdom;
21 Mario Negri Institute for Pharmacological Research, Milan, Italy;
22 Karolinska Institutet, Stockholm, Sweden;
23 German Cancer Research Center, Heidelberg, Germany;
24 Institute of Health and Society, University of Oslo, Oslo, Norway;
25 National Cancer Registry, Cork, Ireland;
26 Formerly International Agency for Research on Cancer, Lyon, France;
27 A. Gemelli University Hospital, Rome, Italy;
28 Wolfson Institute of Preventive Medicine, Queen Mary University of London, United Kingdom;
29 Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Beijing, China;
30 American Cancer Society, Atlanta, Georgia, United States of America;
31 Academic Medical Centre, Amsterdam, the Netherlands;
32 National Health Screening Services, Ministry of Health, Elderly & Community Care, Valletta, Malta;
33 University of Applied Sciences FH Joanneum, Graz, Austria;
34 Catalan Cancer Strategy, L’Hospitalet de Llobregat, Spain;
35 Digestive Cancer Registry of Burgundy, INSERM U866, University and CHU, Dijon, France;
36 Lynn’s Bowel Cancer Campaign, Twickenham, United Kingdom;
37 National Israeli Breast and Colorectal Cancer Detection, Haifa, Israel;
38 Department of Gynecological Pathology and Cytology, University Medical Center Ljubljana, Slovenia;
39 University of Vermont, Burlington, Vermont, United States of America;
40 Cancer Prevention and Research Institute (ISPO), Florence, Italy;
41 National Cancer Centre, Tokyo, Japan;
42 Cancer League of Eastern Switzerland, St. Gallen, Switzerland von Karsa L et al.
43 Cancer Registry of Norway, Oslo, Norway;
44 Telemark Hospital, Skien, Norway;
45 Danish Cancer Society, Copenhagen, Denmark;
46 Hospital General Universitario de Alicante, Alicante, Spain;
47 Maria Sklodowska-Curie Memorial Cancer Centre and Medical Centre for Postgraduate Education, Warsaw, Poland;
48 Lithuanian Cancer Registry, Vilnius, Lithuania;
49 U1086 INSERM - UCBN, CHU Caen, France;
50 The Catholic University of Korea College of Medicine, Seoul, Republic of Korea;
51 St. George’s Hospital, London, United Kingdom;
52 University of Latvia, Riga, Latvia;
53 Oregon Health & Science University, Portland, Oregon, United States of America;
54 University of Copenhagen, Copenhagen, Denmark;
55 MaMMa Healthcare Institute, Budapest, Hungary;
56 Health Administration Central Region Portugal, Aveiro, Portugal;
57 National Public Health Institute, Ljubljana, Slovenia;
58 Gastroenterology Unit, Valduce Hospital, Como, Italy;
59 GlaxoSmithKline Pharma Europe, London, United Kingdom;
60 Regional Health Administration, Coimbra, Portugal;
61 Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America;
62 Prof. Dr Ion Chiricuţă, Cluj-Napoca, Romania;
63 Catalan Institute of Oncology, L’Hospitalet de Llobregat, Spain;
64 University of North Carolina, Chapel Hill, North Carolina, United States of America;
65 Ruhr Universität, Bochum, Germany; 66 Epidemiology and Cancer Registry, Institute of Oncology Ljubljana, Slovenia;
67 University of Toronto and Cancer Care Ontario, Toronto, Canada;
68 Bispebjerg University Hospital, Copenhagen, Denmark;
69 Institut Arnault Tzanck, St Laurent du Var, France;
70 Mount Sinai Hospital, Toronto, Canada;
71 Institute for Cancer Research and Treatment, Candiolo-Torino, Italy;
72 Faculdade de Medicina - Universidade de Coimbra, Coimbra, Portugal;
73 Ministry of Health, Luxembourg, Luxembourg;
74 Cancer Foundation of India, Kolkata, India;
75 The University of Chicago, Department of Medicine, Hematology–Oncology Section, Center for Clinical Cancer Genetics, Global Health, Chicago, United States of America;
76 University Hospitals Coventry & Warwickshire NHS Trust, Coventry, United Kingdom;
77 Charles University and Military University Hospital, Prague, Czech Republic;
78 Chinese Academy of Medical Sciences, Beijing, China;
79 Department of Cancer Screening, Stockholm Gotland Regional Cancer Centre, Stockholm, Sweden;
80 University of Leuven, Leuven, Belgium;
81 Agenzia Sanitaria e Sociale Regionale-Regione Emilia-Romagna, Bologna, Italy;
82 University of Miami, Miami, Florida, United States of America;
83 Niigata University, Niigata, Japan;
84 Memorial Sloan–Kettering Cancer Center, New York, United States of America;
85 Gastrointestinal Services, Flinders University, Adelaide, Australia;
86 State Patient Fund, Vilnius, Lithuania;
87 NHS Endoscopy, Leicester, United Kingdom

Endoscopy 2013; 45: 51-59
Official Organ of the European Society of Gastrointestinal Endoscopy (ESGE) and affiliated societies

Abstract:

Population-based screening for early detection and treatment of colorectal cancer (CRC) and precursor lesions, using evidence-based methods,can be effective in populations with a significant burden of the disease provided the services are of high quality. Multidisciplinary, evidence-based guidelines for quality assurance in CRC screeningand diagnosis have been developed by experts in a project co-financed by the European Union. The 450-page guidelines were published in book format by the European Commission in 2010.They include 10 chapters and over 250 recommendations, individually graded according to the strength of the recommendation and the supporting evidence. Adoption of the recommendations can improve and maintain the quality and effectiveness of an entire screening process, including identification and invitation of the target population, diagnosis and management of the disease and appropriate surveillance in people with detected lesions. To make the principles, recommendations and standards in the guidelines known to a wider professional and scientific community and to facilitate their use in the scientific literature, the original content is presented in journal format in an open-access Supplement of Endoscopy. The editors have prepared the present overview to inform readers of the comprehensive scope and content of the guidelines.


Prevalence of high risk Human Papillomavirus and Cervical Intraepithelial Neoplasias in a previously unscreened population – A pooled analysis from three studies

Partha Basu1, Srabani Mittal2, Suchismita Bhaumik1, Shyam Sunder Mandal1, Ranajit Mandal1, Pradip Das1, Anushree Samaddar1, Chinmayi Ray4, Maqsood Siddiqi3, Jaydip Biswas1, Rengaswamy Sankaranarayanan5

1 Chittaranjan National Cancer Institute, Kolkata, India;
2 Child in Need Institute, Kolkata, India;
3 Cancer Foundation of India, Kolkata, India;
4 Vivekananda Institute of Medical sciences, Kolkata, India;
5 International Agency for Research on Cancer, Lyon, France

International Journal of Cancer , 132, 1693-99, 2013

Abstract:

Introduction:

Population prevalence of Human Papillomavirus (HPV) and Cervical Intraepithelial Neoplasias (CIN) is an important indicator to judge the disease burden in the community, to monitor the performance of cervical cancer screening program and to assess the impact of HPV vaccination program. India being a country without any cervical cancer screening program has no published data on the population prevalence of CIN and only a few large community based studies to report the high risk HPV prevalence. The objective of our study was to study HPV and CIN prevalence in a previously unscreened population.

Methodology:

We pooled together the results of three research studies originally designed to assess the performance of Visual Inspection after Acetic Acid Application (VIA) and Hybrid Capture 2 (HC2). Nearly 60% of the screened women had colposcopy irrespective of their screening test results. The diagnosis and grading of cervical neoplasias were based on histology.

Results:

The age standardized prevalence of HPV by HC2 test was 6.0%. Age-adjusted prevalence of CIN 1 and CIN 2 was 2.3% and 0.5% respectively. The age-adjusted prevalence of CIN3 was 0.4% and that of invasive cancer was 0.2%.

Conclusion:

The prevalence of high risk HPV was relatively low in the population we studied, which is reflected in the low prevalence of high grade CIN. The prevalence of CIN3 remained constant cross age groups due to absence of screening.

Impact:

The prevalence data as a benchmark will be useful to monitor future screening and vaccination program in the study areas