Call for Abstract

25th International Conference on Gastroenterology and Hepatology, will be organized around the theme “Theme: "Innovations in Gastroenterology and Hepatology: Bridging Research with Patient Care"”

Gastroenterology 2025 is comprised of keynote and speakers sessions on latest cutting edge research designed to offer comprehensive global discussions that address current issues in Gastroenterology 2025

Submit your abstract to any of the mentioned tracks.

Register now for the conference by choosing an appropriate package suitable to you.

Non-invasive liver diagnostics have revolutionized the detection and monitoring of liver diseases, offering safer and more accessible alternatives to traditional liver biopsy. Among the most notable advancements is elastography, particularly transient elastography (FibroScan), which measures liver stiffness to assess the degree of fibrosis. This painless method is commonly used for conditions like NAFLD, NASH, and cirrhosis. The advent of Magnetic Resonance Elastography (MRE) has further improved accuracy by combining MRI with elastography, offering detailed images that help assess liver stiffness and architecture.

Additionally, serological biomarkers such as FibroTest and FIB-4 provide blood tests that predict liver fibrosis severity by measuring specific serum markers. These tests are non-invasive, cost-effective, and widely used in clinical practice. Another breakthrough is the development of genetic and molecular biomarkers, which help identify individuals at higher risk for liver diseases based on their genetic profiles.

 

The human microbiome, composed of trillions of bacteria, fungi, viruses, and other microorganisms, plays a crucial role in maintaining overall health, including liver function. The gut-liver axis refers to the bidirectional communication between the gastrointestinal (GI) tract and the liver. The microbiome influences liver health through various mechanisms, including immune modulation, metabolism, and the detoxification process.

  • Gut Microbiome and Liver Disease: An imbalance in the gut microbiome, known as dysbiosis, has been linked to several liver diseases, including non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), cirrhosis, and liver cancer (HCC). Dysbiosis can lead to increased intestinal permeability ("leaky gut"), allowing harmful substances, such as endotoxins, to enter the bloodstream.
     
  • Microbiome in NAFLD and NASH: In the context of NAFLD and NASH, gut microbiota imbalances contribute to the accumulation of fat in the liver, insulin resistance, and inflammatory responses that aggravate liver injury. Studies suggest that microbiome therapies, such as probiotics, prebiotics, and dietary interventions, may improve liver function by restoring microbial balance and reducing inflammation.
     
  • Liver Detoxification and Metabolism: The gut microbiome also affects the liver's role in detoxifying harmful substances. Certain gut bacteria produce short-chain fatty acids (SCFAs) like butyrate, which help maintain gut barrier function and regulate the liver's metabolic processes. The gut microbiota also influences the liver’s ability to metabolize drugs, alcohol, and other toxic substances, making it a key player in detoxification.

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, often associated with chronic liver diseases, such as chronic hepatitis B, hepatitis C, and non-alcoholic fatty liver disease (NAFLD). Due to the liver's ability to compensate for damage, HCC is frequently diagnosed at an advanced stage when treatment options are limited. However, early detection significantly improves prognosis, making timely screening and management essential.

Early detection of HCC is crucial for effective treatment. Screening is particularly recommended for high-risk individuals, such as those with cirrhosis, chronic hepatitis B or C, and other liver diseases. Imaging techniques like ultrasound are commonly used for initial screening, often supplemented by alpha-fetoprotein (AFP) testing, a biomarker that may be elevated in the presence of HCC. Additionally, contrast-enhanced ultrasound, CT scans, and MRI can be used for further evaluation to assess the size, location, and vascular characteristics of the tumor.

Management of HCC:

Once diagnosed, the management of HCC depends on the tumor's size, location, and the underlying liver function. Surgical resection is the most effective treatment for early-stage HCC, providing the best chance for long-term survival. However, many patients are not candidates for surgery due to cirrhosis or advanced liver disease. In these cases, liver transplantation may be considered, especially for patients with early-stage HCC and underlying cirrhosis.

Pediatric gastroenterology and hepatology are medical specialties focusing on the diagnosis and treatment of gastrointestinal (GI) and liver disorders in children. This branch of medicine addresses a broad range of conditions affecting the digestive system, liver, pancreas, and bile ducts in infants, children, and adolescents.

Gastroenterology in pediatrics deals with disorders such as gastroesophageal reflux disease (GERD), irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and celiac disease. Pediatric gastroenterologists diagnose and manage conditions related to chronic abdominal pain, constipation, diarrhea, and malnutrition. They use advanced diagnostic tools like endoscopy, imaging, and biopsy to evaluate and treat these conditions.

Pediatric hepatology, on the other hand, focuses on liver diseases such as hepatitis, liver fibrosis, fatty liver disease (NAFLD), biliary atresia, and genetic liver disorders. Conditions like neonatal cholestasis and cirrhosis are also common concerns in pediatric hepatology. Early diagnosis and treatment are crucial to prevent long-term liver damage or failure.

Pediatric gastroenterology and hepatology require a multidisciplinary approach, often involving nutritionists, surgeons, and other specialists. Advancements in treatments, such as biologic therapies for IBD and liver transplantation, have significantly improved outcomes, enhancing the quality of life for affected children. The field continues to evolve with ongoing research into genetic causes and novel therapies for pediatric GI and liver disorders.

Colorectal cancer (CRC) remains one of the leading causes of cancer-related deaths worldwide. This track focuses on the latest advancements in early detection, diagnostic tools, and therapeutic options for managing colorectal cancer, a disease that can be more effectively treated when caught at an early stage.

Diagnosis:

Once a potential case of CRC is identified, imaging technologies (e.g., CT scans, MRI, and PET scans) and biopsy techniques are crucial for confirming the diagnosis and staging the cancer. Advanced endoscopic procedures allow for more accurate detection of polyps and early-stage tumors. Genetic testing is also playing a significant role in understanding the molecular profile of tumors, guiding treatment decisions, and providing personalized therapeutic strategies.

Therapeutics:

Treatment for colorectal cancer depends on the stage of the disease. Surgical resection remains the cornerstone for early-stage CRC. For more advanced stages, chemotherapy, radiation therapy, and targeted therapies like monoclonal antibodies (e.g., anti-EGFR and anti-VEGF) have shown promising results. Immunotherapies, particularly checkpoint inhibitors like PD-1 and CTLA-4 inhibitors, have revolutionized treatment for some advanced CRC patients. Ongoing research into personalized medicine and novel drug combinations continues to improve outcomes and reduce side effects, offering hope for better long-term survival and quality of life.

The gut-brain connection is a complex, bidirectional communication system between the gastrointestinal (GI) system and the brain. This axis involves the gut microbiome, the enteric nervous system (ENS), and signaling pathways such as the vagus nerve and immune responses. The gut microbiome, which consists of trillions of microorganisms, influences brain function by producing neurotransmitters like serotonin and dopamine, essential for mood and cognitive function.

Dysbiosis, or an imbalance in the gut microbiome, has been linked to various neurological and psychiatric conditions, including anxiety, depression, and cognitive decline. Moreover, individuals with gastrointestinal disorders like irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and functional dyspepsia often experience psychological symptoms such as stress, anxiety, and depression. This highlights the critical role of the gut-brain axis in managing both physical and mental health.

Research in this area is leading to new therapeutic approaches, including psychobiotics (probiotics aimed at mental health), dietary interventions, and mindfulness-based therapies, which aim to restore balance in the gut microbiome and improve both GI and neurological symptoms. Understanding the gut-brain connection offers a novel approach to treating conditions that overlap in both gastrointestinal and mental health domains, ultimately improving patient care.

Precision medicine in gastroenterology involves tailoring treatments for gastrointestinal diseases based on an individual’s genetic, environmental, and lifestyle factors. This approach utilizes genomic testing and biomarkers to identify personalized treatment plans, enhancing therapeutic efficacy and minimizing side effects. For conditions like inflammatory bowel disease (IBD), colorectal cancer, and liver diseases, precision medicine helps pinpoint the best treatment options by analyzing genetic mutations and tumor markers.

Additionally, microbiome research plays a key role in understanding how gut bacteria influence disease progression and treatment response. Pharmacogenomics allows clinicians to select medications based on how a patient’s genetic makeup affects drug metabolism. This personalized approach also extends to diet and lifestyle modifications, offering patients a comprehensive treatment plan. Ultimately, precision medicine improves outcomes, reduces complications, and enables early detection of conditions like colorectal cancer, offering patients more effective, individualized care in gastroenterology.

Advanced gastrointestinal imaging techniques play a pivotal role in the diagnosis, treatment, and monitoring of gastrointestinal (GI) diseases. These methods provide detailed, non-invasive insights into the GI tract, enabling accurate identification of conditions such as inflammatory bowel disease (IBD), gastroesophageal reflux disease (GERD), colon cancer, and liver diseases.

One of the most significant advancements is magnetic resonance imaging (MRI), which offers high-resolution imaging of the liver, pancreas, and other abdominal organs. MRI enterography is particularly useful in assessing small bowel diseases, providing detailed images without the need for radiation. Endoscopic ultrasound (EUS) is another important technique, allowing real-time imaging of the GI tract, pancreas, and surrounding tissues. It’s especially valuable for detecting early stages of cancers and assessing organ involvement in chronic diseases.

Additionally, computed tomography (CT) and positron emission tomography (PET) scans are utilized to evaluate and stage cancers, track disease progression, and guide treatment plans. Capsule endoscopy, which involves swallowing a pill-sized camera, offers a minimally invasive method for visualizing the small intestine and is especially helpful in diagnosing conditions like Crohn’s disease and small bowel tumors.

Recent advancements in molecular biology, genomics, and immunology have accelerated the development of personalized medicine in gastroenterology. Researchers are identifying genetic markers, biomarkers, and immune pathways that can help predict disease progression, such as in inflammatory bowel disease (IBD), hepatitis, and colorectal cancer. These insights enable the development of targeted therapies, improving treatment outcomes and minimizing adverse effects.

Translational research also plays a vital role in understanding the microbiome's role in GI and liver health, leading to innovations in microbiota-based therapies like fecal microbiota transplantation (FMT). Additionally, advancements in stem cell therapy and gene editing show potential in treating liver diseases like cirrhosis and liver cancer.

By integrating basic science with clinical trials, translational research holds the promise of faster, more effective treatments for a wide range of gastrointestinal and hepatological conditions, ultimately improving patient care and outcomes. This track emphasizes the importance of collaboration between researchers, clinicians, and industry to expedite the translation of new discoveries into practice.

Immunotherapy has emerged as a groundbreaking approach in the treatment of gastrointestinal (GI) cancers, including colorectal cancer, esophageal cancer, gastric cancer, and liver cancer. Unlike traditional treatments such as chemotherapy and radiation, which target cancer cells directly, immunotherapy aims to enhance the body's immune system to recognize and attack cancer cells more effectively.

One of the most promising immunotherapies in GI cancer treatment is checkpoint inhibitors. These drugs, such as pembrolizumab and nivolumab, block immune checkpoints like PD-1 and PD-L1, which cancer cells exploit to evade immune detection. By inhibiting these checkpoints, the immune system is activated to target and destroy cancer cells.

Another promising approach is adoptive T-cell therapy, where a patient's own T-cells are extracted, modified, and reintroduced to fight cancer. Cancer vaccines, designed to stimulate an immune response against specific tumor markers, are also being explored.

Immunotherapy has shown significant success in advanced stages of GI cancers, particularly in patients with microsatellite instability-high (MSI-H) or mismatch repair deficiency (dMMR) tumors. These cancers tend to respond well to immune checkpoint inhibitors, offering hope for patients with previously limited treatment options.

Chronic liver disease management focuses on slowing disease progression, preventing complications, and improving quality of life. The approach depends on the specific condition, such as cirrhosis, chronic hepatitis, or fatty liver disease. Early diagnosis through regular blood tests, imaging, and liver biopsies helps monitor disease severity. Lifestyle changes, including weight loss, a healthy diet, avoiding alcohol, and managing associated conditions like diabetes, are crucial for disease control.

Medications, such as antivirals for hepatitis B or C and drugs for fatty liver disease, help reduce inflammation and prevent further damage. Managing complications like ascites, variceal bleeding, and hepatic encephalopathy is vital to improving patient outcomes. For advanced liver disease or cirrhosis, liver transplantation may be considered. Overall, the goal is to halt progression, minimize symptoms, and enhance long-term health, offering patients a more stable and manageable condition.

Liver regeneration and stem cell research have garnered significant attention in recent years due to their potential to revolutionize the treatment of liver diseases, including cirrhosis, hepatitis, and liver cancer. The liver’s remarkable ability to regenerate naturally has inspired research into harnessing this process to treat liver failure and chronic diseases.

Stem cell-based therapies have emerged as a promising strategy for liver regeneration. Scientists are exploring various types of stem cells, including induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and hepatic progenitor cells, to promote liver tissue repair and regeneration. These stem cells can differentiate into functional liver cells, helping to restore liver function in patients with severe liver damage.

Recent advancements in genetic engineering and 3D cell culture models have improved our understanding of liver regeneration at the molecular and cellular levels. Researchers are focusing on identifying the key genes, proteins, and growth factors involved in the regeneration process, aiming to stimulate these pathways to enhance liver healing.

While still in the early stages, advancements in stem cell therapy and liver regeneration offer hope for patients with end-stage liver disease, providing a future where liver transplants may be less necessary and liver function can be restored using cutting-edge regenerative techniques.

Gut-associated lymphoid tissue (GALT) plays a crucial role in maintaining intestinal immunity and protecting the body from pathogens. As an integral part of the mucosal immune system, GALT consists of structures like Peyer’s patches, mesenteric lymph nodes, and isolated lymphoid follicles, which work together to detect and respond to harmful antigens and regulate immune responses in the gastrointestinal (GI) tract.

Recent advances in research have highlighted the dynamic role of GALT in regulating both local and systemic immunity. Studies have demonstrated how GALT is involved in not only defense against infections but also in maintaining immune tolerance to food antigens and the gut microbiota, which is essential for preventing chronic inflammation and autoimmune diseases. A dysregulated GALT can contribute to the development of inflammatory bowel diseases (IBD), food allergies, and even systemic autoimmune conditions.

Innovations in immunomodulatory therapies targeting GALT are emerging as potential treatments for these conditions. For instance, the manipulation of T-cell subsets and regulatory T-cells (Tregs) within GALT could offer new approaches to managing conditions like Crohn’s disease, ulcerative colitis, and irritable bowel syndrome (IBS). Additionally, oral vaccines and probiotic-based interventions are being investigated to harness GALT’s immune responses in promoting better health and preventing disease.

Understanding the complexities of GALT-mediated immune responses holds the key to developing novel therapies for various GI diseases, making it a rapidly advancing field of research with immense potential for improving gastrointestinal health.

Artificial Intelligence (AI) is transforming the field of colorectal cancer (CRC) diagnosis by offering advanced tools for early detection, which is crucial for improving patient outcomes. Early-stage colorectal cancer often presents with subtle symptoms or none at all, making it challenging to detect through traditional methods. AI-driven technologies are being developed to address this gap and provide more accurate and timely diagnoses.

Machine learning algorithms, particularly deep learning, are increasingly being applied to analyze medical imaging data, including colonoscopy and CT colonography. These AI systems can automatically detect and highlight potential abnormalities in images, such as polyps or lesions, which might be missed by the human eye. By assisting radiologists and gastroenterologists, AI can enhance their diagnostic accuracy and speed, leading to earlier intervention and better survival rate.

By integrating AI into clinical workflows, healthcare providers can achieve faster, more precise diagnoses, reducing the time between detection and treatment. The future of colorectal cancer diagnosis lies in combining AI with traditional diagnostic methods.

Artificial Intelligence (AI) is rapidly revolutionizing clinical decision-making by enhancing the ability of healthcare professionals to diagnose, treat, and manage patients with greater accuracy and efficiency. In the field of gastroenterology and hepatology, AI is playing an increasingly important role in decision support, allowing for personalized treatment plans based on comprehensive data analysis.

AI-driven algorithms are used to analyze vast amounts of patient data, including electronic health records (EHRs), imaging studies, lab results, and genetic profiles. These systems can identify patterns and correlations that may not be immediately apparent to human clinicians, offering valuable insights to guide decision-making. For instance, AI tools can help identify patients at high risk for conditions like colorectal cancer, liver cirrhosis, and inflammatory bowel disease (IBD), facilitating earlier interventions and improving patient outcomes.

In clinical decision support, AI is also used to recommend personalized treatment plans based on patient-specific characteristics such as genetics, lifestyle, and co-existing conditions. AI systems can predict the effectiveness of various therapies, optimize drug dosages, and identify potential side effects, ultimately enhancing the precision of treatment.

Incorporating AI into clinical workflows also helps reduce clinician burnout by automating routine tasks, such as data entry and initial data analysis, allowing healthcare providers to focus on more complex patient care aspects. As AI continues to evolve, its role in clinical decision-making will expand, making healthcare more efficient, personalized, and accessible.

Endoscopy has long been a cornerstone in the diagnosis and treatment of gastrointestinal (GI) and hepatobiliary disorders. Recent advancements in endoscopic techniques have significantly improved the accuracy, safety, and effectiveness of procedures, offering patients less invasive alternatives for diagnosis and treatment. In this track, we explore the cutting-edge innovations that are shaping the future of endoscopy and their applications in gastroenterology and hepatology.

One of the most notable advancements is high-definition endoscopy with narrow band imaging (NBI), which provides enhanced visualization of the mucosal surface, enabling earlier detection of abnormalities such as dysplasia, early-stage cancers, and inflammatory lesions. This technology improves the ability to distinguish between benign and malignant lesions, which is particularly beneficial for early colorectal cancer detection and monitoring conditions like inflammatory bowel disease (IBD).

Endoscopic ultrasound (EUS) has also seen significant advancements, combining endoscopy with ultrasound to offer detailed imaging of the GI tract, pancreas, liver, and surrounding structures. EUS-guided fine needle aspiration (FNA) has become an essential tool for diagnosing pancreatic cancer, liver lesions, and lymph node metastases, providing minimally invasive biopsy options.

Finally, AI integration into endoscopy is set to transform the field. Machine learning algorithms can analyze endoscopic images in real-time, helping identify potential areas of concern such as polyps, tumors, and other abnormalities that might be missed by human eyes. AI-powered systems also assist in training and standardizing procedures, ensuring better outcomes and improving procedural efficiency.

Overall, these advancements are improving patient care by enabling earlier detection, less invasive treatments, and faster recovery times, making endoscopy a vital tool in modern gastrointestinal and hepatobiliary medicine.

The human microbiome, which consists of trillions of microorganisms living in and on our body, plays a crucial role in maintaining health and influencing disease development. In recent years, emerging research has uncovered significant links between the microbiome and colorectal cancer (CRC), making it a focus of intensive study in gastroenterology.

Colorectal cancer is one of the most common cancers worldwide, and while genetic factors and lifestyle are well-known contributors, the role of the gut microbiome is gaining increasing attention. The composition of gut bacteria can influence the development of CRC through various mechanisms, such as immune modulation, inflammation, and the metabolism of dietary components.Conversely, a diverse microbiome with a high abundance of protective bacteria, such as those producing butyrate, has been shown to help maintain colon health and may protect against CRC development.

This track will explore the latest research on the gut microbiome’s role in colorectal cancer, including its impact on disease pathogenesis, the potential for microbiome-based diagnostics, and the therapeutic interventions that may arise from this growing field of study. The integration of microbiome research into CRC care has the potential to revolutionize prevention, diagnosis, and treatment, ultimately improving patient outcomes.

Liver transplantation is a life-saving procedure for patients with end-stage liver diseases, but its success depends heavily on managing immune responses and understanding genetic factors. Immunology plays a central role in both the rejection of transplanted livers and the long-term survival of transplant recipients. The body's immune system recognizes the donor liver as foreign and may initiate an immune response, leading to acute or chronic rejection. This requires careful immunosuppressive therapy to prevent rejection while managing the risks of infection and malignancy.

Genetic factors, both in the donor and recipient, influence transplant outcomes. Genetic incompatibility between donor and recipient, particularly with human leukocyte antigens (HLA), can affect the likelihood of rejection. Research into genomic profiling is helping to identify patients at higher risk for rejection or graft failure, allowing for more personalized treatment strategies.

This track will delve into the latest advancements in the immunology and genetic mechanisms influencing liver transplantation, offering insights into improving graft survival, minimizing rejection, and advancing personalized care for transplant patients.

Autoimmune gastrointestinal (GI) disorders, including conditions such as Celiac disease, Inflammatory Bowel Disease (IBD), and Autoimmune Hepatitis, result from the immune system mistakenly attacking the body’s own tissues. These diseases are characterized by chronic inflammation, mucosal damage, and impaired organ function. Understanding the genetic and immunological mechanisms that underlie these disorders is crucial for improving diagnosis, treatment, and patient outcomes.

Genetic susceptibility plays a significant role in the development of autoimmune GI disorders. Specific genetic variants, such as those found in the HLA (Human Leukocyte Antigen) region, have been strongly associated with conditions like Celiac disease and IBD. These genetic factors influence immune system activation, altering how the body responds to environmental triggers like infections or dietary factors.

This track will explore the latest discoveries in the genetic and immunological mechanisms driving autoimmune GI disorders. The goal is to enhance understanding of these conditions, identify novel therapeutic targets, and contribute to the development of more effective, personalized treatments for patients suffering from these chronic conditions.

Non-Alcoholic Fatty Liver Disease (NAFLD) and its more severe form, Non-Alcoholic Steatohepatitis (NASH), are major contributors to liver disease worldwide, with increasing prevalence due to rising rates of obesity, diabetes, and metabolic syndrome. Understanding the genetic pathways involved in the development and progression of NAFLD and NASH is crucial for identifying high-risk individuals, improving diagnostics, and developing targeted therapies.

Genetic factors play a central role in the onset and progression of NAFLD and NASH. Several studies have identified genetic polymorphisms that influence susceptibility to these diseases, particularly genes involved in lipid metabolism, inflammation, and fibrosis. For instance, variants in the PNPLA3 gene have been associated with an increased risk of liver fat accumulation and progression to NASH and cirrhosis. Other genes, such as TM6SF2 and MBOAT7, are also implicated in modulating liver fat storage, inflammation, and fibrosis.

This track will focus on the latest research uncovering the genetic and epigenetic mechanisms that drive the progression of NAFLD to NASH, exploring potential biomarkers for early diagnosis and novel genetic targets for therapeutic intervention. Understanding these pathways will help pave the way for personalized treatment strategies to address this growing global health issue.

CRISPR-Cas9, a groundbreaking gene-editing tool, is revolutionizing the field of gastroenterology and hepatology by offering precise and targeted genetic modifications. In gastrointestinal research, CRISPR is used to investigate genetic mutations responsible for diseases like inflammatory bowel disease (IBD), colorectal cancer, and Celiac disease. By editing genes involved in immune responses and cell proliferation, researchers can uncover the underlying mechanisms of these conditions and develop targeted therapies.

In hepatology, CRISPR is advancing the understanding and treatment of liver diseases such as Non-Alcoholic Fatty Liver Disease (NAFLD), hepatitis, and liver fibrosis. The technology enables researchers to modify specific genes, offering potential for gene therapies aimed at correcting genetic mutations responsible for hereditary liver disorders like Wilson’s disease and alpha-1 antitrypsin deficiency.

This track will explore the latest advancements in using CRISPR-Cas9 for both gastrointestinal and liver diseases, highlighting its potential for genetic disease modeling, therapy development, and regenerative medicine. Furthermore, it will address the ethical and clinical challenges of translating CRISPR-based treatments into real-world applications, bringing us closer to precision medicine in the treatment of complex gastrointestinal and hepatic disorders.

Gastrointestinal inflammation, a hallmark of diseases such as Inflammatory Bowel Disease (IBD), Crohn's disease, and ulcerative colitis, is driven by complex genetic and immune system interactions. Understanding the genetic pathways involved in these inflammatory processes is essential for developing more effective and targeted therapies. This track will focus on the latest advancements in identifying and targeting the specific genetic pathways that contribute to gastrointestinal inflammation.

In addition, the role of epigenetics, microRNAs, and immune system modulation in gastrointestinal inflammation will be explored. Advances in gene editing technologies, such as CRISPR-Cas9, are opening new doors for directly manipulating these genetic pathways, potentially leading to the development of more precise, long-lasting treatments.

This session will highlight the most recent breakthroughs in genetic research related to gastrointestinal inflammation, showcasing novel therapeutic strategies and discussing the challenges and future directions for targeting genetic pathways to improve patient outcomes in GI inflammatory diseases.

Liver diseases such as Non-Alcoholic Fatty Liver Disease (NAFLD), hepatitis, and cirrhosis are often driven by complex inflammatory processes that are influenced by both genetic and environmental factors. Identifying genetic markers that predict inflammatory responses is crucial for early diagnosis, prognosis, and personalized treatment strategies. This track will explore the latest research on genetic biomarkers that can help predict and monitor inflammatory responses in liver diseases.

Specific genes involved in immune regulation, inflammation, and liver cell apoptosis are being studied to understand their role in disease progression. Markers such as IL-6, TNF-alpha, and NLRP3 inflammasome have been implicated in chronic liver inflammation, leading to fibrosis and cirrhosis. Additionally, recent advancements in genomic technologies are enabling researchers to identify single nucleotide polymorphisms (SNPs) and gene expression profiles that are linked to variations in inflammatory responses among individuals.

This session will delve into the latest advancements in genetic markers for liver disease, highlighting their potential for improving clinical outcomes and informing future therapeutic strategies.

MicroRNAs (miRNAs) are small, non-coding RNA molecules that play a crucial role in regulating gene expression at the post-transcriptional level. In the context of liver diseases, miRNAs are emerging as key regulators of various genetic pathways involved in disease progression, inflammation, fibrosis, and tumorigenesis. This track will focus on the growing body of research exploring the role of miRNAs in the genetic regulation of liver diseases such as Non-Alcoholic Fatty Liver Disease (NAFLD), cirrhosis, hepatitis, and hepatocellular carcinoma (HCC).

MiRNAs influence the expression of genes involved in critical processes such as cell proliferation, apoptosis, fibrosis, and immune response. Dysregulation of specific miRNAs has been linked to liver injury, fibrosis progression, and the development of liver cancer. For example, miR-122, a liver-specific miRNA, has been shown to regulate lipid metabolism and is a potential biomarker for liver disease progression.

This session will provide insights into the mechanisms by which miRNAs regulate liver disease pathways and their potential as biomarkers and therapeutic targets in liver disease management.

Hepatitis D, caused by the Hepatitis D virus (HDV), is one of the most severe and often overlooked forms of viral hepatitis. This virus can only infect individuals already infected with Hepatitis B virus (HBV), making it a co-infection that accelerates liver disease progression. Known as the “silent epidemic,” Hepatitis D can lead to chronic liver disease, cirrhosis, and a significantly increased risk of hepatocellular carcinoma (HCC).

This session will focus on the pathophysiology, diagnosis, and management of Hepatitis D, shedding light on the global prevalence and clinical challenges it presents. Special attention will be given to the epidemiology, genetic characteristics, and clinical outcomes associated with HDV infection, particularly in regions with high HBV endemicity.

Discussions will also include current treatment options, such as pegylated interferon and the search for novel antiviral agents. As HDV is often underdiagnosed, the session will emphasize the importance of early detection, screening methods, and public health initiatives. The development of an effective Hepatitis D vaccine remains a priority, and the track will provide the latest updates on vaccine research.

Attendees will gain valuable insights into managing this deadly co-infection and its long-term impact on liver health.

Hepatitis E, caused by the Hepatitis E virus (HEV), is a significant global health issue, primarily transmitted through contaminated water. While often self-limiting, the disease can cause severe complications in pregnant women, immunocompromised patients, and individuals with underlying liver conditions. HEV is gaining attention due to its rising incidence in both developed and developing countries, with emerging reports of genotype 3 and genotype 4 strains in non-endemic regions.

This session will explore the latest research on the epidemiology, molecular mechanisms, and clinical management of Hepatitis E. Key topics will include genetic diversity of HEV, the impact of coinfections with other viruses, and new diagnostic tools.The track will highlight recent advances in vaccine development, as well as antiviral treatments for chronic infections, which remain a challenge due to limited therapeutic options. Researchers will discuss novel drug candidates and immunization strategies to reduce the global burden of HEV.

Attendees will gain a deeper understanding of Hepatitis E, its clinical implications, and the latest advances in diagnosis, treatment, and prevention.

Pancreatitis, an inflammation of the pancreas, can be classified as acute or chronic, both of which present significant challenges in medical management. This track focuses on the latest advances in the treatment of acute and chronic pancreatitis, aiming to improve patient care and outcomes.For acute pancreatitis, early diagnosis and intervention are critical. This session will explore the latest research on early nutritional support, minimally invasive techniques, and drainage procedures for managing pancreatic fluid collections. Additionally, the track will cover the use of biomarkers for predicting disease severity and outcomes, as well as new pharmacological therapies.

In chronic pancreatitis, a key focus will be on pain management, endoscopic procedures, and surgical options for managing complications such as pancreatic duct obstruction. The role of pancreatic enzyme replacement therapy (PERT), endoscopic ultrasound, and the management of autoimmune pancreatitis will also be discussed. Recent developments in stem cell therapy and regenerative medicine to repair damaged pancreatic tissue offer new hope for patients.

This track will provide valuable insights into the latest diagnostic tools, therapeutic strategies, and ongoing research, offering practical takeaways for clinicians managing acute and chronic pancreatitis.

Obesity and bariatric surgery are closely linked, as bariatric surgery is a common treatment option for individuals with severe obesity, particularly when other methods like diet and exercise have failed. Obesity is a chronic condition characterized by excessive body fat, which can lead to a range of health issues, including diabetes, heart disease, hypertension, and sleep apnea.

Bariatric surgery involves surgical procedures that alter the digestive system to help patients lose weight. The most common types include:

  1. Gastric Bypass (Roux-en-Y): This surgery reduces the stomach size and reroutes the small intestine to limit food intake and nutrient absorption, leading to significant weight loss.
  2. Sleeve Gastrectomy: The stomach is reduced in size by removing a large portion, limiting food intake and promoting weight loss.
  3. Adjustable Gastric Banding: A band is placed around the upper part of the stomach to create a small pouch, reducing food intake.
  4. Biliopancreatic Diversion with Duodenal Switch: A more complex procedure that involves both stomach size reduction and rerouting of the small intestine.