Demo
Demo
From Bibliography to Educational Materials
In the evolving landscape of education, artificial intelligence offers a transformative opportunity for teachers and students alike. This demo aims to illustrate how, by leveraging the capabilities of today’s leading AI technologies, it is possible to generate a wide array of high-quality educational resources around a single subject.
The introduction of AI has multiplied the teacher’s capacity for content creation by a factor still difficult to measure. It dramatically enhances not only the number of materials produced but also their quality and variety. By using AI tools, educators can now design courses, synthesize complex bibliographies, create detailed lecture slides, develop case studies, formulate quizzes, and offer personalized learning aids—at a speed and depth never before imaginable.
Our ambition is to show how these tools can support both the teacher and the student: assisting educators in the efficient conception and realization of their courses, and providing students with richer, more diverse materials to facilitate a deeper understanding and assimilation of the subject matter.
Ultimately, rather than replacing the teacher’s voice, AI stands as a powerful ally—enabling educators to spend more time focusing on pedagogical strategy, personal mentorship, and the transmission of clinical wisdom that remains irreplaceable.
Wilms Tumor: From Biology to Personalized Therapeutics
Wilms Tumor:
From Biology to Personalized Therapeutics
Bibliography
WT1 (Wilms Tumor 1) is a zinc finger transcription factor encoded on chromosome 11p13, originally described as a tumor suppressor in pediatric nephroblastoma. However, its function is far more complex: depending on the cellular context, WT1 can act as an oncogene or a tumor suppressor. It regulates gene transcription, cellular differentiation, and organogenesis during development, and maintains homeostasis in adult tissues.
WT1 produces multiple isoforms (+KTS, –KTS) through alternative splicing, each playing distinct roles in gene expression and tumor biology. Its expression is typically restricted to embryonic and select adult tissues like kidney podocytes, but is aberrantly upregulated in a wide array of cancers.
a) Acute Myeloid Leukemia (AML)
WT1 is mutated in ~15% of AML patients and often co-mutated with FLT3-ITD or NPM1. These mutations influence the Wnt/β-catenin, TET2, and MEG3 pathways, promote leukemogenesis, and impact response to chemotherapy. WT1 overexpression enhances stemness, self-renewal, and resistance to apoptosis.
b) Myelodysplastic Syndromes (MDS)
WT1 is overexpressed in ~50% of MDS patients. It is associated with poor prognosis and disease progression. WT1 interacts synergistically with FLT3 mutations, contributing to transformation to AML.
c) T-cell Acute Lymphoblastic Leukemia (T-ALL)
WT1 mutations (~10% of T-ALL cases) activate the IL7R-JAK-STAT pathway and impair TP53-mediated DNA damage response. Co-occurrence with NOTCH1/FBXW7/FLT3 mutations suggests an oncogenic synergy.
d) Chronic Myeloid Leukemia (CML)
WT1 expression is increased in advanced CML via BCR-ABL1 activation. It contributes to resistance against tyrosine kinase inhibitors like imatinib by interfering with apoptosis and gene regulation through the WT1/ZNF224/c-Myc axis.
e) Acute Promyelocytic Leukemia (APL)
WT1 interacts with the PML-RARA fusion protein, altering differentiation. Noncoding mutations in WT1 may also contribute to leukemogenesis.
a) Lung Cancer
WT1 promotes VEGF isoform expression and angiogenesis. It may serve as a target to reverse resistance to EGFR inhibitors in non-small-cell lung cancer (NSCLC).
b) Breast Cancer
WT1 isoforms have distinct roles in ER-positive and triple-negative breast cancer. They affect ERα signaling, promote EMT and metastasis, and may predict resistance to hormone therapy. WT1-targeted vaccines in combination with endocrine therapy show promising early results.
c) Neuroblastoma
In neuroblastoma, WT1 often functions as a tumor suppressor. It promotes differentiation through inhibition of PI3K/Akt and MAPK/ERK pathways. However, high WT1 is paradoxically associated with poor prognosis in non-MYCN-amplified tumors.
d) Prostate Cancer
WT1 downregulates E-cadherin, facilitating migration and metastasis. It also modulates VEGF transcription and androgen receptor activity, influencing hormone resistance.
e) Hepatocellular and Pancreatic Cancer
WT1 influences Wnt/β-catenin, JAK2/STAT3, and NF-κB pathways. It promotes tumorigenesis, invasion, and modulates immune escape mechanisms. In pancreatic ductal adenocarcinoma, it interacts with STAT3 and miR-216a/KRT7, facilitating metastasis.
f) Ovarian Cancer
WT1 is highly expressed in serous subtypes and promotes EMT via ERK1/2 and E-calmodulin pathways. It is involved in mesenchymal–epithelial transitions (MET) and cancer stem cell formation through interaction with EZH2.
g) Astrocytomas/Glioblastoma
WT1 contributes to glioblastoma aggressiveness and poor prognosis. Its inhibition enhances radiosensitivity, and WT1 vaccines elicit cytotoxic immune responses in glioma patients.
a) Chemical Inhibitors
Agents like 17-AAG (Hsp90 inhibitor), TMPyP4 (G-quadruplex stabilizer), and daunorubicin reduce WT1 expression. These agents inhibit tumor growth and increase chemosensitivity in WT1-overexpressing malignancies.
b) Natural Products
Compounds such as bufalin, shikonin, aloin, curcumin, and resveratrol suppress WT1 expression and modulate apoptosis pathways. Marine-derived molecules also show potential in WT1 inhibition.
c) Kinase Modulators
GSK3β affects WT1 stability and degradation. Dasatinib is proposed as a modulator to reduce cytokine release syndrome in combination therapies.
Conclusion and Future Prospects
WT1 is a compelling, dual-role protein in cancer biology. It functions as a context-dependent oncogene or tumor suppressor, modulating multiple pathways in both hematologic and solid tumors. Its overexpression correlates with treatment resistance and poor prognosis.
Targeting WT1 is now a key area of translational research, with therapeutic strategies spanning small molecules, natural products, and immunotherapies. Though many approaches remain in preclinical or early clinical stages, the diversity of mechanisms through which WT1 drives cancer supports its status as a pan-cancer therapeutic target.
Comprehensive Summary
This narrative review explores the evolution of personalized medicine in the context of Wilms tumor (WT), the most common renal malignancy in children. It synthesizes recent advancements in molecular diagnostics, risk stratification, and therapeutic innovations, offering a roadmap for the development of individualized care pathways in pediatric oncology.
Wilms tumor accounts for roughly 7% of all childhood cancers, with a high survival rate (>90%) for localized disease in developed countries. However, variability in outcomes persists due to biological heterogeneity, socioeconomic disparities, and treatment-related toxicity. The shift toward risk-adapted and genetically-informed strategies is positioned as key to improving outcomes while minimizing late effects.
Key genetic players such as WT1, CTNNB1, SIX1, MYCN, and WTX are central to WT pathogenesis. The review emphasizes that:
The authors advocate for integrating imaging, molecular, and epigenetic data to enable early detection and treatment personalization:
The review underscores a paradigm shift from stage-based to biomarker-based stratification:
The review details how radiomic analysis and AI-assisted imaging can predict histological subtypes, therapeutic response, and relapse risk. Coupled with genomic data, they provide the basis for integrative predictive models that will soon guide both surgical planning and post-treatment surveillance.
The authors propose a multi-dimensional predictive framework based on:
Conclusion
This review establishes that the future of WT therapy lies in personalization. By integrating multi-omic diagnostics, predictive modeling, and innovative therapies, clinicians can move toward tailoring care to the individual child. These efforts promise not only higher survival, but also reduced toxicity, better quality of life, and long-term survivorship.
Comprehensive Summary
This retrospective audit analyzes 23 pediatric Wilms’ tumor (WT) cases managed at a single Indian tertiary-care center over a 3-year period (2021–2024). The study evaluates how SIOP Umbrella protocols were applied and adapted in a resource-limited setting, offering insights into protocol adherence, surgical approaches, tumor response, immunohistochemistry, and outcomes in unilateral and bilateral WT.
Summary:
This comprehensive review synthesizes current knowledge on Wilms tumor (WT)—also called nephroblastoma—with an emphasis on its clinical profile, treatment paradigms, molecular markers, and research gaps. It provides a global perspective and addresses both standardized treatment advances and novel, personalized approaches, while also recognizing challenges faced in resource-limited settings.
Wilms tumor is the most common pediatric renal malignancy, accounting for over 90% of childhood kidney tumors. It typically occurs in children under 5 years old, and more than 90% of diagnoses are made before age 3.
Conclusion
The review emphasizes the multifactorial nature of Wilms tumor involving histological, genetic, and environmental components. As survival improves, efforts must focus on risk stratification, reduction of treatment burden, and integration of precision medicine tools. Future success in WT care hinges on international cooperation, advanced molecular diagnostics, and the development of integrative predictive models.
Comprehensive Summary:
This review explores the evolution of prognostic factors in Wilms tumor (WT) management, emphasizing the integration of clinical, histological, and molecular markers to guide risk-adapted therapy. Through comparative analysis of the Children’s Oncology Group (COG) and International Society of Pediatric Oncology (SIOP) protocols, the article outlines how risk stratification has shaped personalized therapy, enhanced outcomes, and reduced treatment toxicity.
Wilms tumor is the most common malignant renal tumor in children and the second most frequent extracranial solid tumor. It occurs in ~1 in 10,000 children and is diagnosed before the age of 5 in the majority of cases. Most tumors are unilateral, but 5–10% are bilateral, often associated with genetic syndromes like WAGR, Denys-Drash, and Beckwith-Wiedemann.
Originally, only tumor stage and histologic subtype were used to define risk and guide treatment. COG and SIOP have progressively expanded these criteria to incorporate tumor volume, genetic markers, response to therapy, and age at diagnosis.
a) Tumor Stage and Histology
b) Loss of Heterozygosity (LOH) at 1p and 16q
c) Chromosome 1q Gain
d) Lung Nodule Response
COG Approach:
SIOP Approach:
Conclusion
The refinement of risk stratification in Wilms tumor has led to safer and more effective therapies, improving survival while reducing long-term toxicity. By incorporating clinical, histologic, and molecular markers, and leveraging treatment response data, clinicians can now offer truly personalized care. Ongoing and future research will focus on refining predictive models and incorporating liquid biopsy, genomic profiling, and AI-based stratification into routine clinical practice.
Extended Summary
This article presents a retrospective analysis of late adverse effects in survivors of Wilms’ tumor (WTS) treated between 1977 and 2023 at a single pediatric oncology center in Spain. With advances in treatment increasing survival rates above 90%, this study focuses on understanding the chronic health conditions that may arise long after therapy completion.
a) Renal Disorders (46%)
b) Cardiac Disorders (23%)
c) Endocrine Disorders (26%)
d) Reproductive Disorders (13%)
e) Second Neoplasms (SN) (9%)
f) Musculoskeletal (28%)
g) Neurological and Pulmonary Disorders (15% each)
h) Skin Disorders (36%), Gastrointestinal (17%), Ophthalmologic (15%), and ORL (28%) effects also observed
3. Psychosocial and Quality of Life Impact
4. Recurrence and Treatment Intensification
5. Conclusions and Recommendations
The study confirms that Wilms tumor survivors are at significant risk for multi-system late effects, particularly affecting renal, cardiac, endocrine, and reproductive systems. It calls for:
These efforts are essential to provide survivors with a quality of life comparable to the general population, acknowledging that treatment side effects may appear decades after remission.
Presentation of the Course Topic
Wilms Tumor (WT), or nephroblastoma, is the most common malignant renal tumor in children, representing a cornerstone of pediatric oncology. Thanks to international collaboration and therapeutic advances, survival rates for localized disease now exceed 90%. However, despite these successes, challenges persist, particularly regarding relapsed, refractory, and high-risk cases, as well as long-term sequelae in survivors.
This course offers an in-depth exploration of Wilms Tumor, beginning with its biological foundations, including the role of key genetic drivers like WT1, CTNNB1, and emerging markers such as 1q gain and TRIM28 mutations. We will review histological subtypes, molecular stratification, and the impact of tumor heterogeneity on prognosis and treatment. A detailed discussion will be dedicated to modern diagnostic approaches, integrating advanced imaging (diffusion-weighted MRI, 3D segmentation) and non-invasive molecular techniques such as liquid biopsy.
Participants will gain a comprehensive understanding of contemporary treatment strategies, from standard protocols based on tumor stage and histology to cutting-edge innovations including immunotherapy, targeted molecular therapies, and nephron-sparing surgery. Special emphasis will be placed on the integration of personalized medicine approaches, where genomic and transcriptomic profiling enables tailoring treatments to individual patient risk profiles.
Finally, the course will address the late effects of Wilms Tumor treatments, such as cardiotoxicity, renal dysfunction, and secondary malignancies, and will outline survivorship care plans adapted to long-term follow-up needs.
Through a multidisciplinary perspective, combining oncology, genetics, radiology, surgery, and survivorship care, this course will equip future urologists and pediatric oncologists with the necessary knowledge and skills to optimize outcomes for Wilms Tumor patients in the era of precision medicine.
Detailed Course Outline with Timed Chapters
1.1 Definition and Historical Background (2 min)
1.2 Epidemiology and Demographics (2 min)
1.3 Clinical and Public Health Relevance (2 min)
1.4 Course Objectives and Structure (2 min)
1.5 Overview of Core Concepts (2 min)
2.1 Histologic Subtypes (5 min)
2.2 WT1 Gene and Key Mutations (10 min)
2.3 Epigenetics and Transcriptional Profiles (5 min)
3.1 Clinical Presentation (3 min)
3.2 Imaging Modalities (5 min)
3.3 Molecular Diagnostics and Liquid Biopsy (5 min)
3.4 Staging Systems (2 min)
4.1 Traditional Prognostic Factors (5 min)
4.2 Molecular Markers and Nomograms (5 min)
4.3 AI and Machine Learning (5 min)
5.1 Standard Treatment Approaches (5 min)
5.2 Nephron-Sparing Surgery (3 min)
5.3 Management of Relapse and Refractory Disease (4 min)
5.4 Emerging Therapeutic Strategies (8 min)
6.1 Structure and Role of WT1 (3 min)
6.2 WT1 Expression in Cancer (3 min)
6.3 Targeting WT1 — Drug Classes (9 min)
7.1 Long-Term Sequelae (5 min)
7.2 Survivorship Protocols (3 min)
7.3 Transition to Adult Care (2 min)
8.1 Integrating Genomics in Therapy (3 min)
8.2 Radiogenomics and Artificial Intelligence (4 min)
8.3 Challenges and Perspectives (3 min)
9.1 Key Learning Takeaways (5 min)
9.2 Clinical Case and Practical Application (5 min)
9.3 Final Q&A (5 min)
Q & A
The primary clinical sign is a painless abdominal mass, often noticed by a parent during bathing or dressing. Other symptoms may include hematuria, hypertension, abdominal pain, fever, and, rarely, respiratory symptoms if lung metastases are present.
The WT1 gene functions as a tumor suppressor crucial for kidney development. Mutations or dysregulation of WT1 can lead to uncontrolled proliferation of nephrogenic cells, contributing to Wilms Tumor pathogenesis, particularly in syndromic cases like WAGR and Denys-Drash syndromes.
COG recommends upfront nephrectomy followed by chemotherapy based on histology and stage. SIOP advocates for preoperative chemotherapy first to shrink the tumor, minimize surgical complications, and then perform nephrectomy. Risk stratification and staging occur differently in both protocols.
LOH at 1p and 16q is associated with a higher risk of recurrence and poorer outcomes. Patients with this genetic alteration typically receive intensified chemotherapy regimens to counteract the increased relapse risk.
Chromosome 1q gain is linked to inferior event-free and overall survival. It is a strong adverse prognostic marker and is being incorporated into future clinical trials for stratifying treatment intensity.
Diffuse anaplastic histology is associated with a poor prognosis due to resistance to chemotherapy and higher relapse rates. It requires more aggressive treatment protocols compared to favorable histology Wilms Tumor.
Ultrasound is the first-line imaging modality for initial diagnosis. MRI, particularly with diffusion-weighted sequences and 3D volumetric segmentation, is preferred for detailed surgical planning and assessing venous involvement.
Liquid biopsies, using cfDNA or ddPCR techniques, detect tumor-specific mutations non-invasively. They offer potential for early diagnosis, real-time treatment monitoring, and relapse prediction without requiring invasive tissue samples.
Late effects include cardiotoxicity from anthracyclines, renal dysfunction, infertility, growth disturbances, and secondary malignancies, especially after radiation therapy. Lifelong surveillance is recommended for survivors
NSS aims to preserve kidney function and is considered in bilateral Wilms Tumors, solitary kidneys, or when preoperative chemotherapy sufficiently shrinks the tumor. It requires careful preoperative imaging and surgical expertise
Emerging therapies include anti-GD2 immunotherapy, HIF-2α inhibitors, Wnt/β-catenin pathway blockers, and WT1-targeted vaccines and small molecules. These are under investigation to improve outcomes in difficult-to-treat cases.
Anaplastic Wilms Tumor requires more aggressive multimodal treatment, including intensified chemotherapy regimens, higher doses of radiotherapy, and sometimes novel agents, given its resistance to standard therapy.
Volumetric assessment via MRI allows precise measurement of tumor shrinkage after chemotherapy, helps surgical planning, predicts histologic response, and is increasingly used in risk-adapted therapy strategies
Personalized medicine aims to stratify patients more accurately based on genetic, molecular, and imaging biomarkers, thereby tailoring therapy intensity to maximize survival while minimizing toxicities and long-term side effects.
Prohibitin (PHB) levels in urine have shown potential as a non-invasive biomarker for early detection of recurrence and prognosis prediction in Wilms Tumor patients. Further validation is ongoing in clinical trials.
Fictitious Clinical Cases
✅ Answer: Immediate nephrectomy followed by histologic staging (COG protocol).
✅ Answer: Intensification with a 3-drug regimen (vincristine, dactinomycin, doxorubicin) plus possible radiotherapy.
✅ Answer: Excellent (>90% 5-year survival).
✅ Answer: WAGR syndrome (Wilms tumor, Aniridia, Genitourinary anomalies, mental Retardation).
✅ Answer: No; irradiation can be omitted if a complete response is achieved.
✅ Answer: Intensified chemotherapy, higher radiotherapy doses, and close monitoring.
✅ Answer: Good histologic response; favorable prognostic indicator.
✅ Answer: Bilateral nephron-sparing surgery if feasible.
✅ Answer: Adjuvant two-drug chemotherapy (vincristine and dactinomycin).
✅ Answer: Early relapse; additional imaging and management should be considered.
✅ Answer: Higher risk of relapse; therapy may need escalation.
✅ Answer: Anthracycline-induced cardiotoxicity
✅ Answer: SIOP protocol (neoadjuvant chemotherapy to reduce tumor size before surgery).
✅ Answer: Much worse; rhabdoid tumors have very poor prognosis.
✅ Answer: Prior chemotherapy (etoposide or alkylating agents) and/or radiotherapy.
MCQ
QUIZ
Synthesis sheet
Standard Treatment
Nephron-Sparing Surgery (NSS)
Emerging Therapies
Iconography and Image Processing
Faire glisser le slider
Faire glisser le slider
Faire glisser le slider
Faire glisser le slider
PowerPoint Slide Deck