Call for Abstract
Scientific Program
22nd Global Summit on Hematology and Blood Disorders, will be organized around the theme ““Cutting-Edge Diagnostics and Therapeutics in Hematology and Blood Disorders””
Blood Disorders 2026 is comprised of keynote and speakers sessions on latest cutting edge research designed to offer comprehensive global discussions that address current issues in Blood Disorders 2026
Submit your abstract to any of the mentioned tracks.
Register now for the conference by choosing an appropriate package suitable to you.
Immunotherapy has emerged as a transformative approach in the treatment of hematologic disorders, harnessing the body’s immune system to target abnormal blood cells and malignancies. Biological mechanisms underlying immunotherapy involve the activation, modulation, or enhancement of immune responses against diseased cells, including leukemic blasts, lymphoma cells, and abnormal plasma cells. Key strategies include monoclonal antibodies, which specifically bind to tumor antigens to induce cell death or recruit immune effectors, and chimeric antigen receptor T-cell (CAR-T) therapy, where patient T cells are genetically engineered to recognize and eliminate malignant cells. Checkpoint inhibitors, another major modality, block inhibitory immune pathways, enhancing anti-tumor immunity. Diagnostic and laboratory methods play a critical role in selecting appropriate patients, monitoring responses, and managing adverse effects. Techniques include flow cytometry to characterize immune cell populations, molecular assays to detect target antigen expression, ELISA and immunoassays to monitor cytokine release, and next-generation sequencing for genetic profiling of malignant cells. Instruments such as automated cell counters, flow cytometers, incubators for cell culture, and gene-editing platforms are essential for immunotherapy development, manufacturing, and monitoring. Clinically, immunotherapy has demonstrated significant success in hematologic malignancies, improving survival rates, achieving remission in refractory cases, and offering curative potential for selected patients. However, challenges such as cytokine release syndrome, immune-related adverse events, and variable patient responses require careful monitoring and management. This session provides delegates with comprehensive knowledge of immune mechanisms in hematology, laboratory techniques and instruments supporting immunotherapy, and clinical applications of immune-based treatments. By understanding these biological principles and technological advances, participants can appreciate the potential of immunotherapy to revolutionize patient care, improve treatment outcomes, and contribute to the ongoing innovation in the field of hematology and blood disorder management.
Advances in hematology research and clinical trials drive innovation in the diagnosis and treatment of blood disorders. Biological research focuses on understanding disease mechanisms, genetic mutations, cellular pathways, and immune responses. Laboratory techniques include flow cytometry, molecular diagnostics, sequencing, biomarker analysis, and functional assays. Instruments such as automated analyzers, high-throughput sequencing platforms, flow cytometers, and imaging systems enhance research accuracy and reproducibility. Clinical trials assess novel therapies, including targeted biologics, gene therapy, CAR-T cells, and stem cell transplantation, evaluating safety, efficacy, and long-term outcomes. This session equips delegates with knowledge of cutting-edge research, laboratory methods, instrumentation, and translational applications, emphasizing the integration of laboratory discoveries into clinical practice to improve patient outcomes and advance hematology globally.
Hematology plays a key role in diagnosing and managing blood-related complications of infectious diseases, including malaria, viral infections, bacterial sepsis, and emerging pathogens. Biological effects include anemia, leukopenia, thrombocytopenia, coagulopathy, and immune dysregulation. Diagnostic methods include complete blood counts, peripheral blood smears, molecular testing, serology, flow cytometry, and coagulation assays. Instruments such as automated hematology analyzers, PCR machines, flow cytometers, and ELISA readers facilitate rapid and accurate detection. Clinical management involves treating the underlying infection, monitoring hematologic parameters, and providing supportive care such as transfusions or anticoagulants. This session provides participants with a comprehensive overview of the interaction between infectious diseases and hematology, laboratory techniques, instrumentation, and clinical management strategies.
Hemostasis and thrombosis research focuses on understanding the balance between blood clotting and bleeding, which is essential for preventing cardiovascular events and treating coagulopathies. Biological studies investigate platelet function, coagulation pathways, fibrinolysis, and endothelial cell interactions. Diagnostic techniques include platelet aggregation tests, coagulation assays, thromboelastography, flow cytometry, and molecular assays to detect clotting factor deficiencies and genetic mutations. Instruments such as coagulation analyzers, flow cytometers, aggregometers, and imaging systems support precise evaluation. Clinical applications include management of thrombotic disorders, bleeding tendencies, and monitoring anticoagulant therapy. This session equips delegates with insights into cutting-edge research, laboratory techniques, and instrumentation in hemostasis and thrombosis, bridging basic science with clinical practice.
Myeloproliferative and myelodysplastic disorders are hematologic conditions characterized by abnormal proliferation or ineffective production of blood cells. Myeloproliferative disorders include polycythemia vera, essential thrombocythemia, and primary myelofibrosis, while myelodysplastic syndromes involve defective hematopoiesis leading to cytopenias and potential progression to leukemia. Biologically, these disorders arise from mutations in hematopoietic stem and progenitor cells, affecting proliferation, differentiation, and apoptosis. Diagnostic methods include complete blood counts, peripheral blood smears, bone marrow biopsy, cytogenetic analysis, flow cytometry, and molecular testing for mutations such as JAK2, CALR, or MPL. Laboratory instruments include automated analyzers, flow cytometers, sequencing platforms, and microscopes. Management strategies involve targeted therapies, cytoreductive treatment, supportive care, and hematopoietic stem cell transplantation. This session provides delegates with an in-depth understanding of the biological mechanisms, diagnostic methods, instrumentation, and therapeutic approaches for myeloproliferative and myelodysplastic disorders.
Transfusion-related complications remain a critical concern in hematology, emphasizing the need for safe practices in blood transfusion and immunohematology. Biologically, complications can arise due to immune reactions, such as hemolytic transfusion reactions, alloimmunization, and graft-versus-host disease, or from infectious agents present in donor blood. Diagnostic and laboratory methods for identifying and preventing these complications include blood typing, crossmatching, antibody screening, direct and indirect Coombs tests, and molecular blood group genotyping. Instruments such as automated blood analyzers, flow cytometers, gel card systems, and ELISA readers are used for precise detection of antigen-antibody interactions, donor compatibility, and pathogen screening. Clinically, prevention strategies include careful donor selection, pathogen inactivation methods, leukoreduction, and vigilant post-transfusion monitoring. Advances in molecular typing, pathogen reduction technologies, and automation have significantly improved transfusion safety. This session equips delegates with comprehensive knowledge of transfusion biology, laboratory techniques, instrumentation, and clinical protocols for preventing and managing complications, ensuring safe, effective, and evidence-based transfusion practices.
Advanced imaging and laboratory technologies are crucial for accurate diagnosis, monitoring, and research in hematology. Biological insights into blood cell morphology, bone marrow structure, and tissue pathology are complemented by imaging modalities such as confocal microscopy, flow cytometry, fluorescence imaging, and automated analyzers. Laboratory techniques include complete blood counts, coagulation testing, molecular diagnostics, and cytogenetic analysis. Instruments like automated hematology analyzers, flow cytometers, high-throughput sequencing platforms, and imaging systems enhance diagnostic precision, reproducibility, and research capabilities. These technologies facilitate early disease detection, characterization of blood disorders, monitoring of therapy response, and translational research. This session highlights the integration of biological understanding, laboratory techniques, and cutting-edge instrumentation, empowering delegates to apply modern hematology technologies in clinical and research settings.
Iron deficiency and nutritional anemias are among the most common hematologic conditions worldwide, caused by insufficient dietary intake, malabsorption, chronic blood loss, or vitamin deficiencies. Biologically, these conditions lead to reduced hemoglobin synthesis, impaired red blood cell production, and oxygen-carrying capacity, resulting in fatigue, pallor, and other systemic symptoms. Diagnostic methods include complete blood counts, peripheral smear examination, serum ferritin, iron studies, vitamin B12 and folate levels, and reticulocyte counts. Instruments such as automated hematology analyzers, spectrophotometers, and ELISA readers are commonly used in laboratories to ensure accurate assessments. Management strategies include dietary supplementation, oral or intravenous iron therapy, and correction of underlying causes. Early and precise diagnosis through modern laboratory techniques ensures effective treatment, prevention of complications, and improved patient outcomes. This session provides delegates with detailed knowledge of the biological basis, diagnostic methods, laboratory instruments, and treatment strategies for iron and nutritional anemias.
Platelet disorders and thrombocytopenia encompass conditions where platelet number or function is impaired, leading to excessive bleeding or clotting. Biologically, these disorders may result from genetic mutations, autoimmune destruction, bone marrow suppression, or medication-induced platelet dysfunction. Diagnostic methods include complete blood counts, peripheral blood smears, platelet aggregation studies, flow cytometry for platelet surface markers, and molecular testing for inherited platelet disorders. Instruments used include automated hematology analyzers, flow cytometers, aggregometers, and microscopes. Clinical management involves platelet transfusions, immunosuppressive therapy, thrombopoietin receptor agonists, and careful monitoring to prevent complications. Understanding the biology and applying precise laboratory methods are essential for effective diagnosis and treatment. This session equips participants with insights into the mechanisms, laboratory evaluation, instrumentation, and therapeutic strategies for platelet disorders.
Gene therapy and genetic engineering are rapidly advancing fields in hematology, offering potential cures for inherited and acquired blood disorders. Biologically, these approaches involve correcting or modifying defective genes responsible for conditions such as sickle cell disease, thalassemia, hemophilia, and immunodeficiencies. Techniques include viral vector-mediated gene delivery, CRISPR-Cas9-based gene editing, and RNA-based therapies, which enable targeted modifications of hematopoietic stem cells or immune cells. Diagnostic and laboratory methods are essential for patient selection, monitoring, and evaluating therapy efficacy. These include molecular genetic testing, sequencing, flow cytometry, PCR assays, and functional studies to assess gene expression and protein production. Instruments such as gene-editing platforms, automated cell culture systems, flow cytometers, PCR machines, and high-throughput sequencing instruments support research and clinical applications. Gene therapy allows personalized treatment by restoring normal gene function, improving hemoglobin production, or enhancing immune function. Clinical outcomes have shown remarkable improvements in patients previously dependent on lifelong transfusions or therapies. Safety monitoring is critical, as off-target effects, immune reactions, and vector-related complications must be managed. This session provides delegates with an in-depth understanding of the biological basis, laboratory techniques, instrumentation, and clinical applications of gene therapy and genetic engineering, emphasizing the transformative potential of these approaches in modern hematology.
Hematologic malignancies are cancers affecting the blood, bone marrow, and lymphatic system, including leukemia, lymphoma, and multiple myeloma. These disorders arise due to abnormal proliferation of blood cells or plasma cells, often caused by genetic mutations, chromosomal translocations, or dysregulated cellular signaling pathways. Leukemia involves overproduction of immature white blood cells, disrupting normal hematopoiesis and causing anemia, infections, and bleeding complications. Lymphoma originates from malignant lymphocytes, affecting lymph nodes and other organs, while multiple myeloma involves clonal expansion of plasma cells in the bone marrow, leading to bone lesions, immunodeficiency, and kidney dysfunction. Diagnostic approaches include complete blood counts, peripheral blood smears, bone marrow biopsy, immunophenotyping via flow cytometry, cytogenetic analysis, and molecular techniques like PCR and next-generation sequencing. Laboratory instruments such as automated hematology analyzers, flow cytometers, fluorescence microscopes, and sequencing platforms ensure accuracy and reproducibility in testing. Therapeutic strategies include chemotherapy, targeted therapy, immunotherapy, monoclonal antibodies, and hematopoietic stem cell transplantation, with emerging treatments such as CAR-T therapy and gene-editing approaches showing promise. The integration of advanced diagnostics with novel therapies enables early detection, precise disease classification, personalized treatment planning, and monitoring of therapeutic responses. This session provides delegates with comprehensive knowledge of the biological mechanisms, state-of-the-art diagnostic tools, laboratory instruments, and innovative treatment options in hematologic malignancies, fostering improved understanding and application in clinical practice.
Clinical research, case studies, and translational hematology focus on applying laboratory discoveries to clinical practice for the diagnosis, treatment, and management of blood disorders. Biological investigations include understanding disease mechanisms, genetic mutations, cellular pathways, and immune responses involved in hematologic conditions. Methods involve laboratory assays, molecular diagnostics, flow cytometry, imaging, and patient monitoring techniques, supported by instruments such as automated analyzers, microscopes, PCR machines, and sequencing platforms. Case studies provide real-world insights into patient management, therapeutic challenges, and treatment outcomes, while translational research bridges basic science findings to clinical applications. This approach enables development of innovative therapies, optimized treatment protocols, and evidence-based strategies for patient care. Delegates gain comprehensive knowledge of research methodologies, diagnostic techniques, laboratory instruments, and clinical applications, fostering collaboration between research and clinical practice and promoting advances in hematology.
Emerging therapies and precision medicine are transforming hematology by enabling personalized approaches to the treatment of blood disorders. Biological understanding of disease mechanisms, genetic mutations, and cellular signaling pathways allows targeted interventions for conditions such as leukemia, lymphoma, hemoglobinopathies, and coagulation disorders. Diagnostic methods include molecular profiling, next-generation sequencing, flow cytometry, and biomarker analysis, using instruments like automated analyzers, sequencing platforms, and high-throughput screening tools. Novel therapies include CAR-T cell therapy, gene editing, monoclonal antibodies, small molecule inhibitors, and targeted biologics, offering improved efficacy and reduced toxicity. Integration of diagnostic data with patient-specific characteristics enables individualized treatment plans, better risk stratification, and monitoring of therapeutic response. This session equips delegates with knowledge of cutting-edge research, advanced instrumentation, and clinical applications of emerging therapies, emphasizing the potential of precision medicine to improve patient outcomes and revolutionize hematology practice.
Rare and autoimmune blood disorders include conditions such as autoimmune hemolytic anemia, aplastic anemia, paroxysmal nocturnal hemoglobinuria, and rare clotting factor deficiencies. These disorders arise from genetic mutations, immune system dysregulation, or unknown environmental triggers, leading to destruction or dysfunction of blood cells, impaired hematopoiesis, and clinical complications such as anemia, thrombosis, or bleeding. Diagnostic approaches include complete blood counts, peripheral blood smears, bone marrow evaluation, flow cytometry, immunoassays, and molecular genetic testing. Laboratory instruments such as automated hematology analyzers, flow cytometers, ELISA readers, and sequencing platforms enable precise detection of abnormal cells, autoantibodies, and genetic defects. Clinical management involves immunosuppressive therapies, targeted biologics, blood transfusions, and stem cell transplantation for severe cases. Understanding the underlying biological mechanisms is essential for accurate diagnosis, therapeutic planning, and research into novel treatments. This session provides delegates with a detailed overview of the biology, diagnostic methods, and instrumentation associated with rare and autoimmune blood disorders, enhancing the ability to recognize, study, and manage these complex conditions in clinical practice.
Blood transfusion and immunohematology involve the study and clinical application of blood component therapy, compatibility testing, and immune-mediated blood disorders. Biologically, transfusion medicine relies on understanding red blood cell antigens, platelet antigens, plasma proteins, and immune responses to ensure safe and effective therapy. Blood disorders requiring transfusion include severe anemia, hemoglobinopathies, clotting factor deficiencies, and blood loss due to trauma or surgery. Diagnostic and laboratory methods include blood typing, crossmatching, antibody screening, direct and indirect Coombs tests, and advanced immunohematology assays. Instruments such as automated blood analyzers, gel card systems, flow cytometers, and serology platforms are essential for accurate detection of antigens, antibodies, and compatibility issues. Clinical applications include red blood cell, platelet, plasma, and cryoprecipitate transfusions, as well as therapeutic apheresis for autoimmune or malignant disorders. Quality control, donor screening, and infection prevention are integral components of safe transfusion practices. Emerging technologies, including molecular blood typing and pathogen reduction techniques, enhance transfusion safety and efficiency. This session equips delegates with an understanding of transfusion biology, laboratory methodologies, instrumentation, and clinical protocols, emphasizing the importance of safe, precise, and effective blood therapy in patient care.
Pediatric hematology focuses on blood disorders in infants, children, and adolescents, which may differ significantly from adult conditions in terms of biology, presentation, and management. Common pediatric blood disorders include congenital anemias, hemoglobinopathies, immune thrombocytopenia, leukemias, lymphomas, and inherited bleeding disorders. Biologically, these conditions arise from genetic mutations, developmental abnormalities in hematopoiesis, immune system dysregulation, or environmental factors affecting blood cell production and function. Diagnostic approaches in pediatric hematology include complete blood counts, peripheral blood smears, bone marrow aspiration and biopsy, hemoglobin electrophoresis, flow cytometry, and molecular genetic testing. Instruments used in laboratories include automated analyzers, microscopes, flow cytometers, and sequencing platforms, allowing precise evaluation of blood cell morphology, immunophenotype, and genetic defects. Clinical management strategies are tailored to the pediatric population, including supportive care, blood transfusions, iron chelation therapy, growth factor administration, chemotherapy, and stem cell transplantation when indicated. Pediatric hematology also emphasizes early detection through newborn screening programs, which help identify hemoglobinopathies and other inherited blood disorders, enabling timely interventions. Advances in gene therapy and targeted molecular treatments offer promising options for treating congenital and inherited hematologic conditions. This session provides delegates with comprehensive knowledge of pediatric blood biology, diagnostic methods, laboratory instruments, and age-specific treatment strategies. By understanding the unique aspects of pediatric hematology, clinicians and researchers can optimize care, improve outcomes, and reduce long-term complications, ensuring better quality of life for affected children and supporting families throughout the treatment journey.
Stem cell and bone marrow transplantation represent transformative therapeutic strategies in hematology, used to treat blood cancers, genetic blood disorders, and bone marrow failure syndromes. Hematopoietic stem cells (HSCs) are multipotent cells capable of self-renewal and differentiation into all blood cell lineages, making them central to restoring healthy hematopoiesis. Transplantation involves collecting HSCs from bone marrow, peripheral blood, or cord blood, followed by conditioning regimens to prepare patients for engraftment. Biological understanding of stem cell niches, immune compatibility, and engraftment mechanisms is critical for successful outcomes. Diagnostic and preparatory methods include HLA typing, flow cytometry for stem cell characterization, colony-forming assays, and molecular testing to assess donor-recipient compatibility and predict complications. Key instruments include automated cell counters, flow cytometers, PCR and sequencing platforms, and cryopreservation systems to store stem cells safely. Clinical applications of transplantation involve allogeneic and autologous approaches, immunosuppressive management, and monitoring for graft-versus-host disease. Emerging techniques, including gene-edited stem cells and CAR-T therapies, expand therapeutic possibilities, offering personalized and targeted treatment options. This session equips delegates with comprehensive knowledge of stem cell biology, transplantation procedures, laboratory methods, and instrumentation, emphasizing their application in clinical practice. Understanding these aspects enhances patient selection, transplantation success rates, and post-transplant care, positioning stem cell therapy as a cornerstone of modern hematology and providing life-saving treatments for patients with complex blood disorders.
Hemoglobinopathies and genetic blood disorders are inherited conditions affecting the structure, function, or production of hemoglobin, red blood cells, or other blood components. Common disorders include sickle cell disease, thalassemia, hereditary spherocytosis, and other congenital anemias. These conditions often result from point mutations, deletions, or gene rearrangements affecting globin chains or red blood cell membrane proteins. Biologically, these mutations lead to hemolysis, ineffective erythropoiesis, chronic anemia, and associated complications such as organ damage and increased susceptibility to infections. Diagnostic methods include complete blood counts, peripheral blood smears, hemoglobin electrophoresis, high-performance liquid chromatography (HPLC), genetic sequencing, and molecular analysis. Laboratory instruments such as automated hematology analyzers, HPLC systems, PCR machines, and sequencing platforms enable precise identification of abnormal hemoglobin variants and genetic defects. Advances in prenatal and neonatal screening allow early detection and timely intervention. Therapeutic approaches include supportive care, blood transfusions, iron chelation therapy, hydroxyurea treatment, and emerging curative strategies such as gene therapy and hematopoietic stem cell transplantation. This session aims to provide participants with a detailed understanding of the molecular and genetic basis of hemoglobinopathies, modern diagnostic techniques and instruments, and clinical strategies for managing these lifelong conditions. Knowledge gained from this session supports accurate diagnosis, improved patient care, and the development of personalized treatment plans for individuals affected by genetic blood disorders.
Coagulation and hemostasis disorders involve abnormalities in the blood clotting process, leading to either excessive bleeding or unwanted clot formation. Hemostasis is a critical physiological process that maintains the balance between bleeding and clotting, involving platelets, coagulation factors, and vascular endothelium. Disorders may include hemophilia, von Willebrand disease, thrombophilia, disseminated intravascular coagulation, and platelet function abnormalities. Biological understanding of these conditions highlights the interplay of coagulation proteins, genetic mutations, and cellular mechanisms affecting clot formation and stability. Diagnostic methods involve coagulation assays such as prothrombin time (PT), activated partial thromboplastin time (aPTT), thrombin generation tests, platelet function tests, and factor activity measurements. Advanced laboratory tools include automated coagulation analyzers, flow cytometers, ELISA readers, and molecular genetic testing platforms. These instruments allow precise assessment of clotting factor deficiencies, platelet abnormalities, and thrombotic tendencies. Understanding coagulation pathways is critical for guiding treatment decisions, including replacement therapies with clotting factors, anticoagulant management, and novel therapies like recombinant factor proteins or gene therapy approaches. This session provides delegates with a comprehensive overview of the biological mechanisms underlying hemostasis and coagulation disorders, the diagnostic techniques and instruments used in laboratories, and emerging therapeutic strategies. By integrating biological knowledge with practical diagnostic applications, participants gain insights into optimizing patient management, preventing complications, and advancing clinical practice in hematology and transfusion medicine.
Blood disorder diagnostics focus on detecting and monitoring hematologic conditions, including anemia, coagulation disorders, hemoglobinopathies, and blood cancers. These disorders may result from genetic mutations, autoimmune reactions, nutritional deficiencies, or disruptions in normal hematopoiesis. Modern diagnostic methods include complete blood counts, peripheral blood smears, reticulocyte counts, coagulation assays, bone marrow evaluation, and molecular genetic testing. Advanced techniques such as PCR, fluorescence in situ hybridization (FISH), next-generation sequencing, and immunophenotyping allow precise identification of abnormal cell populations, genetic mutations, and disease-specific biomarkers. Laboratory instruments like automated hematology analyzers, flow cytometers, microscopes, and sequencing platforms enhance accuracy, reproducibility, and efficiency. Blood biomarkers, including proteins, genetic signatures, and cellular markers, are essential for early disease detection, classification, and monitoring progression. These diagnostic tools are critical for informing clinical decision-making, guiding research studies, and supporting personalized therapeutic strategies. By understanding the biological basis of blood disorders and utilizing advanced diagnostics and instrumentation, clinicians and researchers can improve patient assessment, optimize treatment planning, and advance hematology research globally.

