Scientific Program

Conference Series Ltd invites all the participants across the globe to attend International Conference and Exhibition on Pediatric Oncology and Clinical Pediatrics Toronto, Canada.

Day 1 :

Conference Series Pediatric Oncology 2016 International Conference Keynote Speaker Georgios Stamatakos photo
Biography:

Georgios Stamatakos is a Research Professor of Analysis and Simulation of Biological Systems at ICCS, National Technical University of Athens. He is the Founder and Director of the In Silico Oncology and In Silico Medicine Group. He has proposed the term and the concept of in silico oncology denoting a new clinical trial driven scientific and technological discipline. He has proposed the concept and pioneered the development of the Oncosimulator. Dr Stamatakos is the coordinator of the EU-US large scale integrating research project “CHIC: Computational Horizons in Cancer: Developing Meta- and Hyper-Multiscale Models and Repositories for In Silico Oncology” FP7-ICT-2011-9(600841)

Abstract:

In silico medicine, an emergent scientific and technological discipline based on clinically driven and oriented multiscale biomodeling, appears to be one of the latest trends regarding the translation of mathematical and computational biological science to clinical practice through massive exploitation of information technology. In silico (i.e. on the computer) experimentation for each individual patient using their own multiscale biomedical data (molecular, histological, imaging etc.) is expected to significantly improve the effectiveness of treatment since reliable computer predictions could suggest the optimal treatment scheme(s) and schedules(s) for each separate case. The Oncosimulator is an information technology system simulating in vivo tumor response to therapeutic modalities within the clinical trial context. The major components, the mathematical approaches and techniques and the function of the technologically Integrated Oncosimulator (IOS) and the Hypermodel based Oncosimulator (HOS) developed within then the framework of several large scale European Commission co-funded projects including ACGT, p-medicine and the EU-US project CHIC are outlined. The technology modules include inter alia multiscale data handling, image processing, execution of the code on the grid/cloud and visualization of the predictions. IOS and HOS appear to be the first worldwide efforts of their kind. In the pediatric oncology context a nephroblastoma, a glioblastoma and an acute lymphocyte leukemia oncosimulators are currently undergoing clinical validation within the framework of real clinical trials. Indicative results demonstrating various aspects of the clinical adaptation and validation process are presented. Completion of these processes is expected to pave the way for the ultimate clinical translation of the systems.

Break: Networking & Refreshments: 11:00-11:15 @ Foyer

Keynote Forum

Imke Bartelink

University of California, USA

Keynote: Drug measurements at the pharmacological target site for individualized pediatric cancer treatment

Time : 11:15-12:00

Conference Series Pediatric Oncology 2016 International Conference Keynote Speaker Imke Bartelink photo
Biography:

Imke H. Bartelink completed her PharmD and PhD in clinical pharmacology in pediatric hematopoietic cell transplantation from the Utrecht Medical Center in Utrecht, The Netherlands and postdoctoral studies in integrative pharmacology from the University of California, San Francisco (UCSF). Currently, Dr. Bartelink is in the Clinical Pharmacology Fellowship at UCSF at the Early Phase Clinical Trials Unit. She published more than 20 papers in high impact peer reviewed journals in the areas of pediatric dosing guidelines, pediatric pharmacokinetic-outcome associations and biomarkers of response.

Abstract:

Cancer drug resistance is a consequence of a complex, multidimensional interplay between tumor, its environment and thernhost. Drugs are oft en assumed to distribute relatively homogeneously from plasma to tumor tissues. However, distributionrnof drug in tumors is highly variable and may not correlate with dose or plasma concentrations. Solid tumors are characterizedrnby a complex and unique microenvironment that consists of infi ltrating immune cells, low pH, dense interstitial matrix, highrninterstitial pressure, and abnormal blood and lymphatic vascular structures. Overexpression of drug effl ux transporters suchrnas P-glycoprotein (MDR1/ABCB1), breast cancer resistance protein (BCRP/ABCG2), and transporters of the multidrugrnresistance-associated protein subfamily (MRP/ABCC) may also limit drug penetration in cancer cells or other (off ) targetrncells. Variability in these factors among cancer types may be a contributing factor why translation from adults to pediatricrnpatients often fails. Until recently, assessment of spatial drug distribution in cancer cells or other target cells and clinical implementation ofrnthese data was limited by technical challenges. New technologies such mass spectrometric and radiolabeled drug imagingrnaddress these challenges and illustrate the promise of applying imaging for optimal development and precision dosing andrncan be directly applied in pediatric dosing studies. In the presentation we will present an overview of our current knowledgernof the association between drug penetration in plasma and target (cancer) cells and their eff ect on tumor response and discussrnapproaches for performing measurements of drug uptake at the pharmacological target site in pediatric patients.

  • Pediatric Oncology
    Pediatric Leukemia
    Pediatric Hematology Oncology
    Neuroblastoma in Children
    Advanced Pediatric Oncology Drugs
    Advances in Pediatric Oncology Treatment

Session Introduction

Liqin Du

Texas State University, USA

Title: Cell differentiation and differentiation therapy in neuroblastoma

Time : 12:00-12:30

Speaker
Biography:

Dr. Du received her PhD degree from the University of Kentucky. She completed her postdoctoral training at University of Texas Southwestern Medical Center at Dallas. She is currently an Assistant Professor in the the Department of Chemistry and Biochemistry at Texas State University in USA. She has published more than 20 peer-review papers. Her current primary research interest is neuroblastoma cell differentiation and differentiation therapy, with goals to identify novel genes that control neuroblastoma cell differentiation and to discover new differentiation agents from various sources for treating neuroblastoma.

Abstract:

Neuroblastoma is the most common solid tumor of infancy and the most common extracranial solid tumor of childhood. Neuroblastoma arises from the neural crest cell precursors of the sympathetic nervous system that fails to complete the process of differentiation, which provides the basis for differentiation therapy, a treatment approach to induce the differentiation of the malignant cells and thereby leading to tumor growth arrest. However, only a limited number of differentiation agents is available to treat neuroblastoma, and resistance to current available differentiation agents is common. This highlights the needs to develop new and more effective differentiation agents. My research goals are to identify novel genes that control neuroblastoma cell differentiation and to discover new differentiation agents from various sources for treating neuroblastoma using a functional high-content screening approach that was recently developed in our group. This approach is based on quantification of the morphological differentiation marker of neuroblastoma cell – neurite outgrowth. By exploiting this screening approach, we have identified a group of novel differentiation-inducing microRNA mimics, synthetic oligonucleotides used to raise intracellular levels of microRNAs. These microRNA mimics induce the differentiation of neuroblastoma cells that are both sensitive and resistant to current differentiation agents, showing the promise of developing microRNA-based differentiation therapeutics to treat neuroblastomas that are resistant to current differentiation agents. Besides the work on microRNAs, we are currently expanding the discovery of novel differentiation agents to other drug sources, including natural products and synthetic small molecule compounds.

Speaker
Biography:

Michael Olin has completed his PhD from from the University of Minnesota in 2006 and postdoctoral studies in the department of Medicine. He has dedicated his efforts to developing immunotherapy for brain tumors. He, among others, have utilized tumor cells as vaccine components, demonstrating promising results with minimal toxicity. He has published more than 20 papers in reputed journals.

Abstract:

Despite the extensive use of tumor-derived vaccines for treatment of CNS tumors, the suppressive tumor-bound protein CD200 has been overlooked to date. CD200 is highly expressed in a variety of human tumors. Our preliminary studies detected CD200 on multiple CNS tumors. This introduces a major problem for the development of tumor-based cancer vaccines. We are “shooting ourselves in the foot” by vaccinating patients with cancer vaccines designed to mount an anti-tumor response. CD200 acts as a checkpoint blockade when engaging its receptor CD200R. CD200 upregulates peptidylprolyl isomerase A (PPIA), resulting in immune suppression. CD200 is expressed on endothelial cells within CNS tumor vessels down-regulating T-cell activation. We have developed a competitive inhibitor peptide overcoming CD200-induced immunosuppression. CD200 inhibitor peptide inhibits PPIA upregulation, enhances cytokine production, and significantly enhances survival. In addition, the CD200 inhibitor results in tumor regression and enhanced survival benefit in our canine model. Impact: We are the first to correlate CD200 in brain tumors and tumor-derived vaccines as an inhibitor of immune activation. Our data suggest that we are suppressing the immune system with the same vaccines designed specifically to induce an anti-tumor response. Tumor endothelial expression of CD200 is also a likely reason for escape from native immune surveillance and failure of other immunotherapeutic approaches. We are optimistic that use of our competitive inhibitor peptide against CD200 and anti-CD200R antibody will ultimately lead to the development of novel therapeutics that improve the efficacy of cancer immunotherapy.

Break: Lunch Break: 13:00-13:45 @ Foyer

Elizabeth Algar

Hudson Institute of Medical Research, Australia

Title: A custom gene panel for interrogating paediatric overgrowth disorders and tumour predisposition.

Time : 13:45-14:15

Speaker
Biography:

Associate Professor Elizabeth Algar was awarded a PhD from Griffith University Australia in 1989 and since then has worked in the broad area of cancer genetics with a specific focus on paediatric cancer. She has led research teams at several universities and research institutes in Australia including the University of Queensland and University of Melbourne. She has authored 60 publications and holds several awards for her work. She has been a panel member of several national grant bodies and is regularly called upon to review journal articles and grant applications. She presently holds the positions of Principal Scientist at Monash Health and Assoc Professor at Monash University and the Hudson Institute.

Abstract:

My laboratory is the predominant Australian testing laboratory for paediatric overgrowth disorders associated with increased cancer risk in childhood, including Beckwith Wiedemann syndrome (BWS) and Hemihypertrophy (HH). Cascade testing typically involves SNP microarray, methylation analysis of imprinting centres on 11p15.5 and CDKN1C (P57) mutation screening. Rare point mutations in NSD1, NLRP2, DNMT1 and ZFP57 have been described in BWS and like disorders as well as deletions and insertions within the 11p15.5 imprinting centres IC1 (H19/IGF2) and IC2 (KCNQ1OT1/CDKN1C). Tumour risk is increased in most genetic and epigenetic subtypes of BWS and HH however degree of risk and tumour type varies between groups. Parents of affected children are often understandably anxious to know the recurrence risk for these conditions and as the number of childhood cancer survivors’ increases, the possibility for transmission of a causative mutation is becoming an increasingly important issue. To improve our capacity to detect predisposing mutations in BWS, HH and in the paediatric tumours that have been described in these conditions, we have designed a gene panel comprising 37 genes as well as intergenic regions spanning imprinting centres on 11p15.5 and 11p13. We have used the Haloplex target enrichment system with sequences run on an Illumina MiSeq. We have performed pilot testing to show that the panel has clinical utility and demonstrates excellent sequence coverage of the 11p imprinting centres. Analysis of results to date has revealed novel mutations including OCT-4 binding site disruption in IC1 and subregions of homozygosity.

Nalini Pati

Canberra Hospital, Australia

Title: Autoimmunity and childhood cancers

Time : 14:15-14:45

Speaker
Biography:

Dr Nalini Pati is currently working as a consultant Adult and Paediatric Haematologist in Haematology Oncology in Canberra Hospital, Canberra and Clinical Senior Lecturer at Australian National University Medical School, Canberra, Australia. He has published more than 20 papers in reputed journals and has been serving as an editorial board member of few journal.

Abstract:

Autoimmunity remains an important causative factor in developing several type of childhood cancers, particularly childhood lumphomas and other lymphoproliferative disorder. Also post diagnosis and treatment for any childhood malignancy, an autoimmune disorder may result in 40% of the cases. Hence this association is very complex. There need to develop various guidelines to be able to screen for these autoimmune disorder in cancer survivors or current clear strategie to reduce the risk. This paper will analyze and address the strength of this association more in detail and would reccommend a suitable screening tool.

Ibtisam H Al-Shuaili

Alberta Children's Hospital, Canada

Title: Inflammatory myofibroblastic tumor in children

Time : 14:45-15:15

Speaker
Biography:

Ibtisam Al Shuaili is a Omani radiology fellow currently doing her fellowship in general pediatrics imaging in Alberta Children's Hospital, Calgary, Canada. She graduated from College of Medicine in Sultan Qaboos University in Muscat, Oman 2009. She finished her radiology residency program at Oman Medical Specialty Board in Oman in 2015. She served as chief resident for the last 3 years of the residency. She have research interest with many research projects which were presented and published in international and national medical journals during the residency and medical school.

Abstract:

Introduction: Inflammatory myofibroblastic tumor (IMT) is a relatively new histopathologic term for an entity previously known as inflammatory pseudotumor, which is a rare pseudosarcomatous inflammatory lesion that occurs in the soft tissues of young adults.

Objectives: The purpose of this review is to describe the pathogenesis, natural history, clinical presentation, imaging features, and management of inflammatory pseudo tumor in various locations throughout the body.

Conclusion: Inflammatory myofibroblastic tumor is a rare benign process mimicking malignant processes and has been found in almost every organ system. Radiologists should be familiar with this entity as a diagnostic consideration to avoid unnecessary surgery.

Speaker
Biography:

Tomokazu Matsuura has graduated from Keio University School of Medicine. He is the chief physician of National Hospital Organization Tokyo Medical Center, Department of Nephrology. He is a councilor of Japanese Society of Nephrology and a part-time Assistant Professor at Keio University, Division of Endocrinology, Metabolism and Nephrology.

Abstract:

Long-term nephrotoxicity of ifosfamide is occasionally progressive, and, in such case, there has been no specific treatment to prevent progression. It has been reported that the presence of karyomegalic interstitial nephritis, which is rare type of interstitial nephritis, may be related to ifosfamide-induced nephropathy with poor prognosis and resistant to the immunosuppressive therapy. A 15-year-old boy presented with progressive nephrotoxicity three years after systemic chemotherapy with ifosfamide and cisplatin for the treatment of osteosarcoma. Renal biopsy revealed the severe tubulointerstitial nephritis with tubular atrophy, and focal global and segmental glomerular sclerosis. It also showed tubular epithelial cells with variably sized nuclei, some of which were massively enlarged, abnormal hyperchromatic, irregular shaped, and bizarre-appearing. These morphological changes were suggestive of the histology of karyomegalic interstitial nephritis. Corticosteroid retarded the progression of nephrotoxicity. The present case is the first report suggesting that corticosteroid was effective against the late-onset renal toxicity by ifosfamide therapy. Our case also suggests that karyomegalic interstitial nephritis may be the result of long-term nephrotoxicity of ifosfamide. Since concurrent treatment with cisplatin is one of the risk factors for ifosfamide nephrotoxicity, there is a possibility that cisplatin may have a synergetic effect with ifosfamide for producing karyomegalic interstitial nephritis.

Speaker
Biography:

Gehan Lotfy has completed her MD at the age of 35 years from Minia University and pediatric oncology board at 2015. She is the director of pediatric hematology and oncology unit. She had published more than 10 papers in reputed journals and had shared in many international pediatric hematology and oncology congresses. She is an Egyptian pediatric board trainer since 2011.

Abstract:

Background: Occult hepatitis B virus infection (OBI) is a form of the disease which does not present with Hepatitis B surface antigens (HBsAg) in the serum of patients; but, HBV-DNA is detectable in the serum and hepatocytes. OBI is an important risk factor to induce post transfusion hepatitis (PTH), liver cirrhosis (LC), hepatocellular carcinoma (HCC) and reactivation of the HBV.

Objective: To detect OBI in frequently blood and blood product transfused pediatric patients.

Patients & Methods: Forty five patients randomly selected from blood transfusion unit in the central blood bank were included. Their ages were 3-18 year. Another known hepatitis B positive age and sex matched patients were enrolled as controls. Hepatitis B surface antigen (HBsAg), anti hepatitis B surface antibodies (antiHBsAb), anti hepatitis B core antibodies (antiHBcAb) and hepatitis B DNA (PCR) were done for both patients and controls.

Results: HBV-DNA; detected by nested PCR; was present in 27 (60%) of the 45 patients of the studied group who were negative for HBsAg. HBcAb was detected in 13(48%) patients from the 27 HBV-DNA positive patients whom were considered as seropositive OBI subjects and 14 patients (52%) were negative and were considered as seronegative OBI subjects.

Conclusions: The potential risk of acquiring occult hepatitis B virus infection is higher in patients receiving multiple and frequent blood transfusions.

Break: Networking & Refreshments: 16:15-16:30 @ Foyer
Speaker
Biography:

After completion of Duquesne Marilyn master thesis at UMONS, She obtained a PhD grant co-funded by Prof. Joelle Nortier, Head of the department of dialysis and kidney transplantation at Erasme hospital in Belgium, and Prof. Jean-Marie Colet, Head of the Human biology & Toxicology laboratory, UMONS. This collaboration also includes the nephrology department of Zagreb University Hospital headed by Dr Bojan Jelakovic. Some results of this current work have been published in the Journal of Cancer Science & Therapy or presented during International conference on traditional and alternative medicine and the kidney week of the Amercican Society of Nephrology.

Abstract:

The composition of a large number of low molecular-weight endogenous molecules fluctuates in human biological fluid according to the physiopathological status of an individual. The spectroscopic analysis of these small molecules in diverse biofluids is called “metabonomic” and generates profiles that can be further associated with specific pathologies.
Approximately 2,000 to 3,000 metabolites are relevant for early clinical diagnostics and most of them are specific for a particular biochemical pathway or patho-chemical processes.
Taken together, subsets of those metabolites constitute functional fingerprints which can be useful in many clinical applications, including pediatry. Clinical metabonomic has already proved its utility in neonatalogy or children's medicine to predict prematurity, mode of delivery, perinatal asphyxia or neurological, kidney and respiratory diseases. Moreover, to better apprehend drugs response from adults to children, pharmacometabonomic is developed to predict drug efficacy or adverse effects and, consequently, can be used to define a safe and effective pediatric dose. Examples from the literature will be presented.
Blood and urine are the most common biofluids used in metabonomics. The simplicity, safety and non-invasive collection of urine samples makes metabonomic a very appropriate diagnostic tool in pediatric medicine.
In addition, in house data obtained in a metabonomic study conducted in the context of a large-scale human kidney disease, the Balkan Endemic Nephropathy, will be presented to demonstrate the potential of this recent omics technology to highlight the mechanism of disease development or to identify biomarkers of diseases.

Speaker
Biography:

Dr. Divya Subburaj Currently doing FNB( Fellowship under the national board) in pediatric hematology and oncology at Apollo hospital,Chennai from feb 2015. She Completed her undergraduation in 2009 from Bangalore medical college and research institute. She have done my specialisation in Pediatrics(M D ) from the above Institute. She Completed MRCPCH part 1 & 2.

Abstract:

A total of fifty four children had received induction chemotherapy consisting of duanorubicin, cytarabine and etoposide as per the UKMRC AML induction protocol were included in the study. Forty seven episodes of febrile neutropenia were recorded. Thirty four percent had culture positive gram negative sepsis. Fifty five percent of the febrile neutropenic episodes were blood culture negative. Enterocolitis was the most common focus of infection in these children. Over the last 3 years (2012-2015) the incidence of gram negative sepsis had risen to 38% when compared to 24% during the 2003 to 2011 period. Though the mortality rates had remained the same in both groups, the morbidity rates which included duration of hospital stay, need for pediatric intensive care support, the use of colistin due to carbapenam resistant infections and the use of granulocyte transfusions to help tide over the sepsis had dramatically increased.

Speaker
Biography:

Dr. Soad K. Al Jaouni is a Professor & Consultant of Hematology and Professor/Consultant of Pediatric Hematology/Oncology, Senior Researcher at Hematology Department, Faculty of Medicine, King Abdulaziz University Hospital (KAUH) a tertiary care medical center, King Abdulaziz University (KAU), Jeddah, Kingdom of Saudi Arabia.

Abstract:

Background: Recent studies showed that Phoenix dactylifera palm date have good nutritious value and medicinal potential.The aim of the study was to determine the effect of regular intake of Phoenix dactylifera palm date called Ajwa on number of infections, hospitalization associated with fever neutropenia and mortality of pediatric cancer patients admitted to King Abdul-Aziz University Hospital, a tertiary care medical center, Faculty of Medicine, King Abdul-Aziz University, Jeddah, Kingdom of Saudi Arabia.

Methods: A non-randomized controlled intervention study was done during the period of 2008-2015. A total of 55 pediatric cancer patients with hematologic or non-hematologic malignancies (who were treated by chemotherapy/radiotherapy) were enrolled in the study. A total of 32 patients were given Ajwa and 23 did not administer Ajwa. Th e outcomes were compared between both groups within the 5 years follow-up period. Culture and sensitivity was done. Th e study was approved by Ethics Research Committee of the hospital.

Results: The study included 27 males and 28 females, with male to female ratio of almost 1:1. Th eir mean age was 9.3 with S.D. of ± 4.4. Children enrolled in the Ajwa group showed minimal positive cultures as compared to non Ajwa (control) groups during the 5-year follow-up period. There is marked reduction in the mortality rate of patients enrolled in Ajwa group as compared to other group (RR=0.74; 95 % CI: 0.06-1.00). Furthermore, there was a signifi cant reduction in the number of hospital admission per year in the Ajwa group (5.74±5.8 times) as compared to others (17.34±10.29 times), with a statistical
signifi cant diff erence (p≤0.05). Additionally, the majority of patients on Ajwa group are currently off treatment. The main cause of death of patients in the non Ajwa group was disease progression and infections (76.9%). Ten patients with Acute Myeloid Leukemia in the Ajwa group (31.2%) showed protection against chemotherapy induced cardiac complications, while this didn't occur among the control group.

Conclusions: Regular intake of Phoenix dactylifera (Ajwa) showed a signifi cant decrease in the number of infections, number of hospitalization per year and mortality rate among pediatric cancer patients. Ajwa have some sort of cardiac protection. Adding Ajwa to the standard treatment of pediatric cancer patients can improve their treatment outcome.

Speaker
Biography:

Dr. Du received her PhD degree from the University of Kentucky. She completed her postdoctoral training at University of Texas Southwestern Medical Center at Dallas. She is currently an Assistant Professor in the the Department of Chemistry and Biochemistry at Texas State University in USA. She has published more than 20 peer-review papers. Her current primary research interest is neuroblastoma cell differentiation and differentiation therapy, with goals to identify novel genes that control neuroblastoma cell differentiation and to discover new differentiation agents from various sources for treating neuroblastoma.

Abstract:

Neuroblastoma is the most common solid tumor of infancy and the most common extracranial solid tumor of childhood. Neuroblastoma arises from the neural crest cell precursors of the sympathetic nervous system that fails to complete the process of differentiation, which provides the basis for differentiation therapy, a treatment approach to induce the differentiation of the malignant cells and thereby leading to tumor growth arrest. However, only a limited number of differentiation agents is available to treat neuroblastoma, and resistance to current available differentiation agents is common. This highlights the needs to develop new and more effective differentiation agents. My research goals are to identify novel genes that control neuroblastoma cell differentiation and to discover new differentiation agents from various sources for treating neuroblastoma using a functional high-content screening approach that was recently developed in our group. This approach is based on quantification of the morphological differentiation marker of neuroblastoma cell – neurite outgrowth. By exploiting this screening approach, we have identified a group of novel differentiation-inducing microRNA mimics, synthetic oligonucleotides used to raise intracellular levels of microRNAs. These microRNA mimics induce the differentiation of neuroblastoma cells that are both sensitive and resistant to current differentiation agents, showing the promise of developing microRNA-based differentiation therapeutics to treat neuroblastomas that are resistant to current differentiation agents. Besides the work on microRNAs, we are currently expanding the discovery of novel differentiation agents to other drug sources, including natural products and synthetic small molecule compounds.

Giannoula Klement

Tufts University School of Medicine, USA

Title: Future Paradigms for Precision Oncology
Speaker
Biography:

Giannoula Lakka Klement, MD, specializes in the treatment of rare tumors and vascular anomalies, and conducts basic science research into tumor and wound angiogenesis, with emphasis on the role of platelets in angiogenesis. A Pediatric Hematologist/Oncologist at Floating Hospital for Children at Tufts Medical Center, she is also the Director of Rare Tumors and Vascular Anomalies Center at Floating Hospital. Dr. Klement is an assistant professor at Tufts University School of Medicine. She is board certified in both Pediatrics and Pediatric Hemaotololgy/Oncology.

Abstract:

Research has exposed cancer to be a very heterogeneous disease with a high degree of intertumoral and intra-tumoral variability. Each individual harbors a unique tumor profile, and this cancer molecular signature makes the use of histology-based treatments problematic. These diagnostic categories, while necessary for care, thwart the use of molecular information for treatment as many molecular characteristics cross tissue type. This difficulty is compounded by the struggle to keep abreast the scientific advances made in all fields of science and by the enormous challenge to organize, cross-reference, and apply molecular data for patient benefit. In order to supplement the site-specific, histology-driven diagnosis with genomic, proteomic and metabolomics information, a paradigm shift in diagnosis and treatment of patients is required. Most physicians are open and keen to use the emerging data for therapy. But even those physicians versed in molecular therapeutics are overwhelmed with the amount of available data, and the lack of tools to integrate it. It is not surprising that even though The Human Genome Project was completed thirteen years ago, our patients have not benefited from the information. Physicians cannot, and should not be asked to process the gigabytes of genomic and proteomic information on their own in order to provide patients with safe therapies. The following consensus summary identifies the needed for practice changes, proposes potential solutions to the present crisis of informational overload, suggests ways of providing physicians with the tools necessary for interpreting patient specific molecular profiles, and facilitates the implementation of quantitative precision medicine.

Annick Beaugrand

International Network for Cancer Treatment and Research, Brazil

Title: New age for pediatrics Oncology?
Speaker
Biography:

Annick Beaugrand Graduated in Medicine from the Federal University of Rio Grande do Norte (2003), Medical Residency in Pediatrics by HUEC (2006) Specialization in pediatric oncology at the Federal University of Parana (PR / Brazil-2008) and the Institut Gustave Roussy (France- 2008 -2011), Degree from the University Paris XI (Paris / France, 2009) in child oncology. Currently a doctor in the House of support to children with cancer and substitute teacher at the Pediatric Department of the Federal University of RN – Brazil and Pediatric Professor of Federal University of RN, Brazil.

Abstract:

Childhood cancer is a success story of modern medicine in which effective treatments have been identified for previously untreatable diseases. Treatment toxicity continues to be substantial despite advances in supportive care, and while survival rates have improved, cures for many with high risk and metastatic disease are not achievable despite aggressive surgical, chemotherapeutic and radiotherapy combination. The cure rates in pediatric oncology have been improved due to standardized treatment strategies and centralization of therapy. Much progress has been made using chemotherapeutic agents and treatment modalities introduced decades ago with refinement to improve disease-free survival. Better understanding of treatment-related toxicity has guided the design of less-toxic therapies. Elucidation of the principles of tumor biology and the development of novel laboratory technologies have led to significant progress, as bringing immunotherapies to the bedside. Understanding the molecular basis are changing the landscape of molecular genetic and genomic testing that may be used to identify risk factors, although the clinical utility of such testing is unclear. Molecular subtyping is instrumental towards selection of model systems for fundamental research in tumor pathogenesis (as new medulloblastoma disease classification) and clinical patient assessment. Can also be used to identify variants that influence drug metabolism or interaction of a drug with its cellular target, allowing customization of choice of drug and dosage. There has been significant progress in the clinical development of monoclonal antibodies as cancer therapies with promising results emerging from pediatric studies. Dramatic progress in scientific discovery and technology has led to rapid development and translation of therapies for the clinic. The challenge to the field of pediatric oncology is to develop biologic based approaches that enhance the benefits of standard therapies, lessen toxicity, and extend the gains in survival to those high-risk groups that have not benefited from standard treatment.

Speaker
Biography:

Michael Olin has completed his PhD from from the University of Minnesota in 2006 and postdoctoral studies in the department of Medicine. He has dedicated his efforts to developing immunotherapy for brain tumors. He, among others, have utilized tumor cells as vaccine components, demonstrating promising results with minimal toxicity. He has published more than 20 papers in reputed journals.

Abstract:

Despite the extensive use of tumor-derived vaccines for treatment of CNS tumors, the suppressive tumor-bound protein CD200 has been overlooked to date. CD200 is highly expressed in a variety of human tumors. Our preliminary studies detected CD200 on multiple CNS tumors. This introduces a major problem for the development of tumor-based cancer vaccines. We are “shooting ourselves in the foot” by vaccinating patients with cancer vaccines designed to mount an anti-tumor response. CD200 acts as a checkpoint blockade when engaging its receptor CD200R. CD200 upregulates peptidylprolyl isomerase A (PPIA), resulting in immune suppression. CD200 is expressed on endothelial cells within CNS tumor vessels down-regulating T-cell activation. We have developed a competitive inhibitor peptide overcoming CD200-induced immunosuppression. CD200 inhibitor peptide inhibits PPIA upregulation, enhances cytokine production, and significantly enhances survival. In addition, the CD200 inhibitor results in tumor regression and enhanced survival benefit in our canine model. Impact: We are the first to correlate CD200 in brain tumors and tumor-derived vaccines as an inhibitor of immune activation. Our data suggest that we are suppressing the immune system with the same vaccines designed specifically to induce an anti-tumor response. Tumor endothelial expression of CD200 is also a likely reason for escape from native immune surveillance and failure of other immunotherapeutic approaches. We are optimistic that use of our competitive inhibitor peptide against CD200 and anti-CD200R antibody will ultimately lead to the development of novel therapeutics that improve the efficacy of cancer immunotherapy.

Speaker
Biography:

Dr Nahla Mobark is pediatric oncologist in Pediatric Hematology & Oncology Department in Cancer Center King Fahad Medical City KFMC, Riyadh Saudi Arabia which is huge tertiary hospital in with around 1000 bed capacity. She had MBBS from KASER-ELAINI Medical College Cairo University, then pediatric residency in Children hospital king Saud medical complex KSMC Ministry of Health (MOH) of Saudi Arabia ,she had membership of Royal College Of Pediatric & Child Health, UK MRCPCH, and then pediatric hematology oncology fellowship program with training in bone marrow transplant. She had got a big experience in diagnosing &management of pediatric patients with hematological malignancies and solid tumours also benign hematology cases i.e .SCA, thalassemia etc. Dr Nahla have been involved with teaching of undergraduate and postgraduate students and nurses throughout her career.

Abstract:

Treatment advances in pediatric Hodgkin’s lymphoma HL have progressed to the point that most children and adolescents diagnosed with Hodgkin’s disease will enjoy long-term disease free survival, although it was the fourth most common malignancy in Saudi children as reported in Saudi cancer registry less information is available about pediatric Non-Hodgkin Lymphoma and its outcome in Saudi Arabia. Pediatric Oncology department is one of the major sections of comprehensive Cancer Centre in KFMC which start accepting new cases of pediatric neoplasm including non-Hodgkin lymphoma in 2006 and now it is consider second center in Saudi Arabia which provide comprehensive care for children with cancer

Nalini Pati

Canberra Hospital, Australia

Title: Autoimmunity and childhood cancers
Speaker
Biography:

Dr Nalini Pati is currently working as a consultant Adult and Paediatric Haematologist in Haematology Oncology in Canberra Hospital, Canberra and Clinical Senior Lecturer at Australian National University Medical School, Canberra, Australia. He has published more than 20 papers in reputed journals and has been serving as an editorial board member of few journal.

Abstract:

Autoimmunity remains an important causative factor in developing several type of childhood cancers, particularly childhood lumphomas and other lymphoproliferative disorder. Also post diagnosis and treatment for any childhood malignancy, an autoimmune disorder may result in 40% of the cases. Hence this association is very complex. There need to develop various guidelines to be able to screen for these autoimmune disorder in cancer survivors or current clear strategie to reduce the risk. This paper will analyze and address the strength of this association more in detail and would reccommend a suitable screening tool.

Speaker
Biography:

Dr Nahla Mobark is pediatric oncologist in Pediatric Hematology & Oncology Department in Cancer Center King Fahad Medical City KFMC, Riyadh Saudi Arabia which is huge tertiary hospital in with around 1000 bed capacity. She had MBBS from KASER-ELAINI Medical College Cairo University, then pediatric residency in Children hospital king Saud medical complex KSMC Ministry of Health (MOH) of Saudi Arabia ,she had membership of Royal College Of Pediatric & Child Health, UK MRCPCH, and then pediatric hematology oncology fellowship program with training in bone marrow transplant. She had got a big experience in diagnosing &management of pediatric patients with hematological malignancies and solid tumours also benign hematology cases i.e .SCA, thalassemia etc. Dr Nahla have been involved with teaching of undergraduate and postgraduate students and nurses throughout her career.

Abstract:

Treatment advances in pediatric Hodgkin’s lymphoma HL have progressed to the point that most children and adolescents diagnosed with Hodgkin’s disease will enjoy long-term disease free survival, although it was the fourth most common malignancy in Saudi children as reported in Saudi cancer registry less information is available about pediatric Non-Hodgkin Lymphoma and its outcome in Saudi Arabia. Pediatric Oncology department is one of the major sections of comprehensive Cancer Centre in KFMC which start accepting new cases of pediatric neoplasm including non-Hodgkin lymphoma in 2006 and now it is consider second center in Saudi Arabia which provide comprehensive care for children with cancer

Annick Beaugrand

International Network for Cancer Treatment and Research, Brazil

Title: New age for pediatrics Oncology?
Speaker
Biography:

Annick Beaugrand Graduated in Medicine from the Federal University of Rio Grande do Norte (2003), Medical Residency in Pediatrics by HUEC (2006) Specialization in pediatric oncology at the Federal University of Parana (PR / Brazil-2008) and the Institut Gustave Roussy (France- 2008 -2011), Degree from the University Paris XI (Paris / France, 2009) in child oncology. Currently a doctor in the House of support to children with cancer and substitute teacher at the Pediatric Department of the Federal University of RN – Brazil and Pediatric Professor of Federal University of RN, Brazil.

Abstract:

Childhood cancer is a success story of modern medicine in which effective treatments have been identified for previously untreatable diseases. Treatment toxicity continues to be substantial despite advances in supportive care, and while survival rates have improved, cures for many with high risk and metastatic disease are not achievable despite aggressive surgical, chemotherapeutic and radiotherapy combination. The cure rates in pediatric oncology have been improved due to standardized treatment strategies and centralization of therapy. Much progress has been made using chemotherapeutic agents and treatment modalities introduced decades ago with refinement to improve disease-free survival. Better understanding of treatment-related toxicity has guided the design of less-toxic therapies. Elucidation of the principles of tumor biology and the development of novel laboratory technologies have led to significant progress, as bringing immunotherapies to the bedside. Understanding the molecular basis are changing the landscape of molecular genetic and genomic testing that may be used to identify risk factors, although the clinical utility of such testing is unclear. Molecular subtyping is instrumental towards selection of model systems for fundamental research in tumor pathogenesis (as new medulloblastoma disease classification) and clinical patient assessment. Can also be used to identify variants that influence drug metabolism or interaction of a drug with its cellular target, allowing customization of choice of drug and dosage. There has been significant progress in the clinical development of monoclonal antibodies as cancer therapies with promising results emerging from pediatric studies. Dramatic progress in scientific discovery and technology has led to rapid development and translation of therapies for the clinic. The challenge to the field of pediatric oncology is to develop biologic based approaches that enhance the benefits of standard therapies, lessen toxicity, and extend the gains in survival to those high-risk groups that have not benefited from standard treatment.