Call for Abstract

11th International Conference on Central Nervous System Disorders & Therapeutics, will be organized around the theme “Neuroprecision Therapeutics: Advancing Brain Repair, Molecular Targeting & Intelligent CNS Care”

CNS 2026 is comprised of keynote and speakers sessions on latest cutting edge research designed to offer comprehensive global discussions that address current issues in CNS 2026

Submit your abstract to any of the mentioned tracks.

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

Rapid advances in neuroscience have enabled the emergence of neuroprecision medicine, a transformative discipline focused on tailoring therapeutic interventions to the molecular, genetic, and physiological signatures of central nervous system disorders. This approach integrates high-resolution biomarker profiling, neural pathway mapping, and individualized drug response prediction to deliver therapies with enhanced accuracy and minimized side effects. Molecular targeting plays a pivotal role, enabling interventions that modulate specific ion channels, neurotransmitter pathways, receptor systems, and gene expression patterns involved in neurological dysfunction. By leveraging patient-specific genomic landscapes, clinicians can identify molecular vulnerabilities such as channelopathies, synaptic dysregulation, or protein abnormalities and apply treatments that directly modulate these mechanisms. The integration of AI-guided predictive tools further enhances decision-making by forecasting therapeutic outcomes and identifying optimal regimens. Neuroprecision medicine holds promise across epilepsy, multiple sclerosis, neurodegeneration, psychiatric disorders, and rare genetic syndromes. As the field advances, combining molecular therapies with regenerative strategies and neural modulation technologies is expected to establish a new era of highly personalized CNS care.

Artificial intelligence is reshaping how neurological disorders are detected, classified, and monitored, providing unprecedented speed and accuracy in clinical decision-making. Machine learning algorithms can analyze massive datasets from brain imaging to electrophysiology, genomics, speech patterns, and digital behavioral markers—to reveal subtle abnormalities undetectable by human evaluation alone. Predictive neuroanalytics models can forecast disease progression, identify early risk states, track subclinical changes, and predict therapeutic response trajectories, transforming early diagnosis and proactive management. Deep-learning systems enhance MRI and PET interpretation, while real-time AI tools embedded in wearable neurotech enable continuous neurological monitoring outside clinical settings. These technologies support faster diagnosis of Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, stroke, epilepsy, and psychiatric disorders by identifying unique digital biomarkers and neural signatures. Moreover, AI-driven platforms are essential for precision stratification in clinical trials, improving patient selection and treatment matching. As neuroanalytics becomes more integrated with personalized medicine, AI will become a core component of intelligent CNS care.

Restoring synaptic integrity and neural circuitry is central to reversing functional decline in neurodegenerative and neurological disorders. Contemporary research focuses on understanding how synaptic pruning, neurotransmitter imbalance, dendritic spine loss, and disrupted plasticity contribute to impaired communication between brain regions. Breakthroughs in synaptic biology are enabling the development of therapies that stimulate dendritic regrowth, enhance long-term potentiation, and promote synaptic resilience. Neuroregeneration strategies include molecular modulators, neurotrophic factors, stem-cell–derived neuronal replacement, extracellular vesicles, and engineered biomaterials designed to encourage axonal repair. Advances in circuit-level mapping using connectomics, optogenetics, and neuromodulation allow precise targeting of dysfunctional pathways and enable restoration of lost or impaired functions. Combining regenerative therapeutics with neural interface technologies such as deep brain stimulation, transcranial magnetic stimulation, and closed-loop neurodevices enhances the possibility of re-establishing healthy brain networks. This field paves the way for restoring cognition, motor control, and sensory processing in disorders like spinal cord injury, stroke, Alzheimer’s disease, traumatic brain injury, and movement disorders.

Neuroinflammation is increasingly recognized as a key driver in the onset and progression of CNS disorders, emerging from complex interactions between neurons, glial cells, and peripheral immune systems. Dysregulated microglial activation, cytokine storms, and chronic inflammatory cycles can accelerate neurodegeneration and impair neural repair. Understanding the mechanisms of immune crosstalk such as blood–brain barrier permeability changes, peripheral immune infiltration, and maladaptive complement activation has opened pathways for targeted immunotherapeutic strategies. Novel treatments aim to modulate neuroinflammatory cascades, reprogram microglial activity, block damaging cytokines, and enhance protective immune pathways. Immunotherapeutics, including monoclonal antibodies, small-molecule inhibitors, RNA-targeted therapies, and nanocarriers, offer precision modulation of inflammatory mediators. These approaches are reshaping therapies for multiple sclerosis, Alzheimer’s disease, autoimmune encephalitis, neuropathic pain, and traumatic brain injury. By targeting both acute and chronic inflammatory processes, next-generation interventions aim to slow disease progression, protect neuronal function, and promote long-term neurological recovery.

The integration of multi-omics technologies has revolutionized the understanding of neurological disorders by revealing how gene expression, epigenetic modifications, and transcriptomic patterns influence disease onset, severity, and therapeutic response. Genomic profiling identifies inherited and acquired mutations underlying conditions like epilepsy, neuromuscular diseases, neurodevelopmental syndromes, and neurodegeneration. Epigenomic studies uncover how environmental factors, stress, aging, and inflammation alter DNA methylation, histone modification, and chromatin architecture, shaping long-term neural behavior. Transcriptomic analysis offers real-time insight into gene activity across neuronal and glial populations, uncovering pathway dysregulation and novel therapeutic targets. These technologies support precision diagnostics, risk prediction, patient stratification, and personalized treatment plans. Furthermore, linking multi-omics datasets with computational modeling enables deeper mechanistic understanding of complex CNS disorders. As sequencing becomes more accessible, multi-omics integration will define a new generation of neurotherapeutics tailored to molecular signatures rather than symptom-based classifications.

Modern neuropharmacology is rapidly advancing with the development of innovative CNS-targeted therapeutics designed to overcome challenges such as the blood–brain barrier, off-target effects, and limited bioavailability. New classes of drugs including small molecules, antisense oligonucleotides, peptides, monoclonal antibodies, and gene therapies are engineered to modulate neural signaling, restore cellular balance, and counteract disease-specific molecular abnormalities. Next-generation drug delivery systems utilize nanoparticles, liposomes, focused ultrasound, and intranasal routes to enhance brain penetration and ensure targeted distribution. These technologies enable precise drug localization, reduced systemic toxicity, and improved therapeutic efficacy. Mechanism-based pharmacology integrates pharmacokinetics, pharmacodynamics, and neural circuit biology to tailor treatments for epilepsy, depression, Parkinson’s disease, rare CNS disorders, and neuro-oncology. As personalized drug optimization becomes increasingly data-driven, advanced neuropharmacology offers a strategic pathway to safer, more effective neurological treatment paradigms.

The study of protein misfolding and aggregation remains central to understanding neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. Abnormal accumulation of proteins including amyloid-β, tau, α-synuclein, and TDP-43 disrupts synaptic signaling, triggers inflammation, and leads to neuronal death. Research continues to unravel the mechanisms driving aggregation, impaired proteostasis, mitochondrial dysfunction, oxidative stress, and lysosomal system failure. Innovations in molecular pathway mapping allow targeted disruption of toxic aggregates, stabilization of native protein structures, and enhancement of proteolytic clearance mechanisms. Novel therapeutics, including immunotherapies, molecular inhibitors, antisense RNA agents, and gene-based interventions, aim to prevent aggregation, neutralize toxic species, and preserve neuronal integrity. Understanding proteinopathy dynamics is essential for developing disease-modifying treatments and slowing progression across multiple neurodegenerative conditions.

The brain–gut–microbiome axis plays a crucial role in regulating neurological function, influencing behavior, immunity, stress responses, and neuroinflammation. Microbial metabolites, neurotransmitter-like molecules, and immune mediators interact with neural pathways through vagal signaling, systemic circulation, and hormonal mechanisms. Dysbiosis of the gut microbiome has been linked to disorders such as autism spectrum disorder, depression, Parkinson’s disease, Alzheimer’s disease, multiple sclerosis, and neuropathic pain. Research demonstrates that modulating the microbiome through probiotics, prebiotics, dietary interventions, microbial metabolites, and fecal microbiota transplantation can impact neuroimmune balance, strengthen gut barrier function, and influence neurochemical pathways. Systems biology approaches help decode microbial signatures associated with disease phenotypes, enabling the development of microbiome-informed diagnostics and therapeutics. As microbiome science evolves, integrating gut-targeted therapies with CNS interventions offers a promising avenue for holistic neurological care.

Stem cell-based therapies and neural engineering technologies are reshaping approaches to CNS repair by enabling regeneration of damaged neural tissue and restoration of compromised function. Pluripotent stem cells, neural progenitors, and engineered cell lines can differentiate into functional neurons, oligodendrocytes, and astrocytes, providing replacement strategies for lost or dysfunctional cells. Bioengineered scaffolds, 3D neural constructs, and biomaterials support cell survival, promote axonal growth, and guide circuit reformation. Neural engineering introduces tools such as optogenetics, bioelectronic implants, and biosignal-responsive devices that interact directly with neural pathways to enhance restoration. These strategies hold immense promise for treating spinal cord injury, stroke, neurodegeneration, and genetic neurological disorders. Advancements in safety, immune compatibility, and long-term integration continue to bring the field closer to clinical translation, establishing regenerative neurobiology as a frontier of future CNS therapeutics.

Precision psychiatry is revolutionizing mental health care by integrating biomarkers, neuroimaging signatures, genetic risk factors, and digital behavioral analytics to understand individual variations in psychiatric disorders. This evidence-driven approach enables accurate diagnosis, prediction of treatment response, and design of personalized therapeutic plans for depression, bipolar disorder, schizophrenia, anxiety disorders, and neurodevelopmental conditions. Digital phenotyping using smartphone sensors, speech patterns, activity data, and cognitive assessments provides real-time behavioral and emotional tracking. Biomarkers such as inflammatory markers, neurotransmitter metabolites, and connectivity patterns offer deeper insight into pathophysiology. Therapeutic modulation includes tailored pharmacology, neuromodulation techniques, brain stimulation, and AI-guided therapy models. Precision psychiatry aims to overcome trial-and-error treatment approaches by adopting a scientific, individualized framework that improves clinical outcomes and long-term mental well-being.

Modern neuroimaging technologies provide unparalleled ability to visualize the structure, function, and connectivity of the human brain. High-resolution MRI reveals anatomical changes, white matter integrity, and microstructural abnormalities, while PET imaging captures metabolic activity, amyloid deposition, neuroinflammation, and receptor dynamics. Connectomics maps complex neural networks, enabling understanding of communication pathways disrupted in CNS disorders. Functional imaging tools, including fMRI and diffusion tensor imaging, allow real-time tracking of brain activity patterns and connectivity changes. These technologies enhance diagnosis, treatment planning, and monitoring across neurodegenerative diseases, epilepsy, psychiatric disorders, traumatic injury, and developmental conditions. The integration of imaging with AI analytics and multimodal biomarkers is propelling neuroimaging toward precision-guided neurological care.

Movement disorders such as Parkinson’s disease, dystonia, essential tremor, and Huntington’s disease arise from dysfunction in basal ganglia circuits, motor cortex pathways, and cerebellar networks. Understanding these neurocircuit dynamics enables development of targeted therapeutic interventions that restore motor control and reduce functional impairment. Innovations include dopamine pathway modulation, deep brain stimulation, focused ultrasound thalamotomy, gene therapies, and circuit-specific neuromodulation. Research continues to uncover the neural mechanisms underlying tremor generation, bradykinesia, rigidity, and dystonic postures. Integrating neural biomarkers, electrophysiological mapping, and real-time motor analytics supports advanced treatment personalization. Emerging strategies also focus on disease modification through neuroprotective compounds, mitochondrial-supportive agents, and anti-inflammatory modulators, offering hope for long-term improvement in movement disorders.

Pediatric CNS disorders represent a highly diverse spectrum of genetic, developmental, metabolic, and neurodegenerative conditions affecting infants and children. Advances in genomic technologies allow precise identification of rare mutations, channelopathies, metabolic errors, and chromosomal anomalies underlying many pediatric neurological disorders. Early recognition of developmental delays, neuromuscular abnormalities, epilepsy syndromes, and neurobehavioral phenotypes is essential for effective intervention. Modern therapies incorporate gene editing, antisense oligonucleotides, metabolic correction strategies, targeted pharmacology, and advanced rehabilitation approaches tailored to pediatric needs. Neurodevelopmental plasticity offers unique opportunities for early therapeutic impact, making timely diagnosis and precision-guided treatment critical. Multidisciplinary care models including neurology, genetics, physiotherapy, behavioral sciences, and nutrition support broader functional development and improved long-term quality of life.

Traumatic brain injury and stroke remain leading causes of long-term neurological disability worldwide, driving demand for advanced neurorehabilitation technologies and therapeutic innovations. Research focuses on understanding acute injury mechanisms such as excitotoxicity, inflammation, oxidative stress, and vascular compromise while developing interventions that preserve neuronal integrity during the critical early window. Rehabilitation strategies emphasize neuroplasticity, encouraging rewiring of brain circuits through targeted exercises, robotic-assisted therapies, virtual reality rehabilitation, and brain–computer interface training. Neurorestorative compounds, stem cell therapies, and noninvasive neuromodulation techniques enhance recovery by promoting synaptic strengthening and functional reorganization. Personalized rehabilitation programs based on imaging markers, functional assessments, and digital tracking provide adaptive pathways for recovery. Continued progress in rehabilitation science aims to restore mobility, cognition, and independence following CNS trauma.

Wearable neurotechnologies and brain–computer interfaces (BCIs) are transforming neurological care by enabling continuous monitoring, real-time neural interaction, and personalized therapeutic modulation. Wearable EEG systems, neuroelectronic sensors, and mobile cognitive platforms capture brain activity, detect abnormal patterns, and support early interventions in epilepsy, sleep disorders, cognitive decline, and mood disorders. BCIs provide direct communication pathways between the brain and external devices, supporting motor restoration, assistive communication, and rehabilitation. Intelligent CNS care models integrate cloud-based analytics, AI decision support, and digital therapeutics to deliver proactive and remote management strategies. These technologies empower clinicians to tailor interventions based on dynamic physiological data while enabling patients to participate actively in their care. As neurotech continues to evolve, it promises to redefine accessibility, precision, and continuity of neurological health management.