While Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases is a rapidly advancing field, many challenges have recently been overcome thanks to the advancement of cell-based technologies in 2024, making these treatments available at DrStemCellsThailand‘s Anti-Aging and Regenerative Medicine Center of Thailand. Ongoing research, particularly in clinical trials around the world, continues to refine Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases, exploring their potential to revolutionize the treatment of various neurological conditions. This progress highlights the promise of harnessing stem cells and bioactive molecules to repair damaged neural tissues and restore function in patients suffering from disorders such as Alzheimer’s disease, Parkinson’s disease, and spinal cord injuries.
The human brain, with its intricate network of billions of neurons, is the seat of consciousness, cognition, and identity. Yet, it is also vulnerable to injury, disease, and degeneration, leading to devastating neurological disorders ranging from Alzheimer’s disease to traumatic brain injury. Despite significant advances in neuroscience, the quest to repair and regenerate damaged brain tissue remains a formidable challenge.
At DrStemCellsThailand‘s Anti-Aging and Regenerative Medicine Center of Thailand, enter our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases. Neural stem cells hold the key to brain repair and regeneration, offering hope for restoring lost function and improving the quality of life for patients facing neurological challenges[1-5].
In the pursuit of understanding the regenerative capacity of neural tissue using Cellular Therapy and Stem Cells for Brain Spinal Cord and Neurodegenerative Diseases, researchers have turned to nature’s marvels for inspiration. Among these wonders is the axolotl (Ambystoma mexicanum), a fascinating amphibian renowned for its extraordinary ability to regenerate lost or damaged body parts, including its brain.
The axolotl’s brain regeneration capabilities are nothing short of miraculous. Following injury, these creatures can mobilize neural stem cells to the site of damage, triggering a complex series of cellular processes that culminate in the regeneration of functional brain tissue.
Unlike humans and many other mammals, which often face permanent brain damage in the wake of injury or disease, axolotls possess the remarkable ability to restore lost neural circuitry and regain normal brain function.
Studies of axolotl brain regeneration have unveiled insights into the dynamic interplay of molecular and cellular mechanisms underlying this process. From the activation of quiescent neural stem cells to the orchestration of axonal growth and synaptic reformation, the axolotl’s regenerative prowess offers a wealth of knowledge with profound implications for human neuroscience and regenerative medicine.
By harnessing the lessons learned from the axolotl’s regenerative abilities, scientists aim to develop novel therapeutic strategies for repairing the injured or diseased human brain.
From promoting endogenous neural stem cell activation to engineering stem cell-based therapies, the potential applications of axolotl-inspired regenerative approaches hold promise for transforming the landscape of neurological care.
In this exploration of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases, we embark on a journey inspired by nature’s ingenuity and fueled by the quest for innovation. By bridging the gap between basic research and clinical translation, we strive to unlock the secrets of brain regeneration and pave the way towards a future where neurological disorders are no longer insurmountable barriers to human well-being[1-5].
Neurodegenerative diseases, including Alzheimer’s, Parkinson’s, Huntington’s, and ALS, significantly impact global health. Alzheimer’s disease alone affects an estimated 50 million people worldwide, while Parkinson’s disease affects over 6 million. Huntington’s disease, although rarer, has a profound impact on affected individuals and their families. ALS, although less prevalent, is a devastating condition with a median survival time of 2-5 years from diagnosis. These diseases not only result in significant morbidity and mortality but also place a substantial burden on healthcare systems and society as a whole, highlighting the urgent need for innovative and effective treatments and interventions[6-10].
The brain consists of various cell types, each with its own specialized function. Understanding these lays a foundation for our specialized protocols of Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases. Here is a list of major brain cell types:
1. Neurons: These are the primary cells responsible for transmitting electrical and chemical signals in the brain. They process and transmit information through synapses.
2. Glial cells (Neuroglia): Glial cells provide support and protection to neurons. There are several types of glial cells, including:
a. Astrocytes: These cells maintain the chemical environment around neurons, provide structural support, and contribute to the blood-brain barrier.
b. Oligodendrocytes: Oligodendrocytes produce myelin, a fatty substance that wraps around neuronal axons and facilitates faster signal transmission.
c. Microglia: Microglia are the immune cells of the brain, responsible for clearing debris, pathogens, and damaged cells through phagocytosis.
d. Ependymal cells: These cells line the brain’s ventricles and assist in producing and circulating cerebrospinal fluid (CSF).
3. Pericytes: Pericytes are cells found in the blood vessels of the brain. They play roles in regulating blood flow, maintaining the blood-brain barrier, and supporting vascular function.
4. Endothelial cells: These cells line the inner surface of blood vessels, including those in the brain. They contribute to the blood-brain barrier and help regulate the exchange of substances between blood and brain tissue.
5. Choroid plexus epithelial cells: These cells are located in the choroid plexus, which produces cerebrospinal fluid (CSF). Choroid plexus epithelial cells contribute to the composition and circulation of CSF.
These cell types work together to ensure the proper functioning of the brain, including information processing, neural communication, maintenance of homeostasis, and protection against external threats[11-15].
Despite significant advancements in medical science, neurodegenerative diseases present several challenges, including the progressive degeneration of neurons leading to cognitive decline, motor dysfunction, and loss of independence. These diseases are characterized by complex and heterogeneous pathologies involving protein misfolding, aggregation, and neuronal death, making treatment development challenging. Additionally, the blood-brain barrier restricts the delivery of therapeutic agents to the central nervous system, further complicating treatment strategies.
The multifactorial nature of neurodegenerative diseases necessitates personalized approaches to diagnosis and treatment, which are still in the early stages of development[16-20].
Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases, specifically utilizing neural stem cells, is undergoing investigation as a promising strategy for regenerating brain, spinal cord, and nerve cells while also potentially attenuating the progression of neurodegenerative diseases. Neural stem cells possess the capacity to differentiate into various neural cell types and integrate into damaged neural circuits, facilitating tissue repair. Additionally, they secrete neurotrophic factors and modulate the inflammatory response, promoting neuroprotection and fostering an environment conducive to neuronal survival. Clinical studies are underway to assess the safety and efficacy of neural stem cell transplantation in conditions such as Parkinson’s disease, Alzheimer’s disease, spinal cord injury, and amyotrophic lateral sclerosis (ALS), with encouraging preliminary results suggesting the therapeutic potential of this approach in mitigating neurodegeneration and improving neurological function[16-20].
– Focus: Utilizing neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) to differentiate into dopamine-producing neurons and integrate into basal ganglia circuitry.
– Mechanisms: Trophic support, neurogenesis, and neuroinflammation modulation.
– Clinical Implications: Stem cell-based therapies for neuronal replacement, ongoing transplantation trials.
– Focus: Using neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) to restore cognitive function by differentiating into mature neurons.
– Mechanisms: Neurotrophic factor secretion, synaptic plasticity promotion, and neuroinflammation modulation.
– Clinical Implications: Stem cell therapies for cognitive decline mitigation, ongoing transplantation trials.
– Focus: Neural stem cells differentiation (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) into medium spiny neurons within the striatum.
– Mechanisms: Neuroprotection through growth factors secretion, excitotoxicity modulation.
– Clinical Implications: Stem cell treatments to delay symptom onset, promising preclinical results.
– Focus: Neural stem cells differentiation (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) into motor neurons and spinal cord incorporation.
– Mechanisms: Trophic support, neuroinflammation modulation, axonal regeneration promotion.
– Clinical Implications: Investigation of stem cell transplantation as a potential therapy, ongoing safety and efficacy trials.
– Focus: Investigating the potential of cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) to modulate neural circuitry and improve social communication skills and repetitive behaviors.
– Mechanisms: Enhancing synaptic connectivity, promoting neuroplasticity.
– Clinical Implications: Improving social interaction, reducing behavioral symptoms[21-34].
– Focus: Exploring cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for promoting cerebellar circuitry repair and enhancing motor coordination.
– Mechanisms: Differentiation into cerebellar neurons, trophic support for existing neurons.
– Clinical Implications: Improving gait, balance, and coordination.
– Focus: Assessing the potential of cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for promoting neural regeneration and reducing spasticity.
– Mechanisms: Differentiation into various neural cell types, modulation of inflammatory responses.
– Clinical Implications: Enhancing motor function, reducing muscle stiffness[21-34].
– Focus: Investigating cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) to promote neurovascular repair and reduce neuronal damage.
– Mechanisms: Promotion of angiogenesis, neuroprotection.
– Clinical Implications: Improving neurological deficits, preventing secondary complications.
– Focus: Exploring cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for neuroregeneration and blood flow restoration.
– Mechanisms: Differentiation into neurons, secretion of neurotrophic factors.
– Clinical Implications: Enhancing functional recovery, reducing disability.
– Focus: Assessing the potential of cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for improving cerebral blood flow and preventing ischemic injury.
– Mechanisms: Neuroprotection, modulation of vascular function.
– Clinical Implications: Reducing the risk of recurrent strokes, improving neurological outcomes[21-34].
– Focus: Investigating cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) to enhance synaptic connectivity and reduce neurodegeneration.
– Mechanisms: Synaptic remodeling, neurotrophic factor secretion.
– Clinical Implications: Improving cognitive function, slowing disease progression.
– Focus: Exploring cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for modulating neuronal excitability and reducing seizure frequency.
– Mechanisms: Inhibition of epileptic activity, promotion of neural circuitry stability.
– Clinical Implications: Seizure control, improvement in quality of life[21-34].
– Focus: Assessing the potential of cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for promoting cognitive development and enhancing synaptic plasticity.
– Mechanisms: Differentiation into neural cell types, enhancement of learning and memory.
– Clinical Implications: Improving cognitive abilities, enhancing adaptive skills.
– Focus: Investigating cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for modulating immune responses and promoting remyelination.
– Mechanisms: Immunomodulation, secretion of growth factors.
– Clinical Implications: Reducing relapse rates, slowing disease progression.
– Focus: Exploring cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for enhancing autonomic function and reducing motor symptoms.
– Mechanisms: Neuroprotection, promotion of neurogenesis.
– Clinical Implications: Improving autonomic dysfunction, enhancing quality of life[21-34].
– Focus: Assessing the potential of cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for promoting muscle regeneration and reducing fibrosis.
– Mechanisms: Differentiation into muscle precursor cells, secretion of growth factors.
– Clinical Implications: Improving muscle strength, delaying disease progression.
– Focus: Investigating cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for enhancing neuromuscular junction function and reducing muscle weakness.
– Mechanisms: Trophic support for muscle fibers, promotion of motor neuron survival.
– Clinical Implications: Improving motor function, enhancing mobility[21-34].
– Focus: Exploring cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for promoting axonal regeneration and reducing inflammation.
– Mechanisms: Differentiation into neuronal and glial cell types, secretion of neurotrophic factors.
– Clinical Implications: Improving sensory and motor function, enhancing quality of life.
– Focus: Assessing the potential of cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for promoting motor neuron survival and muscle strength.
– Mechanisms: Trophic support for motor neurons, promotion of neuromuscular junction integrity.
– Clinical Implications: Improving motor function, extending lifespan[21-34].
– Focus: Investigating cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) for promoting spinal cord regeneration and reducing inflammation.
– Mechanisms: Modulation of immune responses, promotion of axonal growth.
– Clinical Implications: Improving sensory and motor deficits, enhancing functional outcomes.
The exploration into cellular therapy and neural stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) in each of these conditions involves understanding their potential mechanisms of action and clinical implications to develop targeted therapeutic interventions that can effectively address the underlying pathology and improve patient outcomes[21-34].
Transplanted neural stem cells and progenitor stem cells (as part of our Cellular Therapy and Stem Cells for Brain, Spinal Cord and Neurodegenerative Diseases) contribute to brain, spinal cord, and nerve repair and remodeling primarily through several mechanisms:
1. Differentiation: Neural stem cells have the ability to differentiate into various neural cell types, including neurons, astrocytes, and oligodendrocytes. This differentiation process allows them to replace damaged or lost cells and contribute to the restoration of neural tissue structure and function.
2. Secretion of Trophic Factors: Neural stem cells secrete various trophic factors, such as brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), and glial cell line-derived neurotrophic factor (GDNF). These factors promote the survival, growth, and function of existing neurons, enhance synaptic connectivity, and stimulate neurogenesis and axonal regeneration.
3. Modulation of Inflammation: Neural stem cells have immunomodulatory properties and can modulate the inflammatory response in the central nervous system. They can suppress harmful inflammatory processes, reduce the activation of microglia and astrocytes, and promote an anti-inflammatory environment conducive to tissue repair and regeneration.
4. Formation of Neuroprotective Microenvironment: Transplanted neural stem cells create a neuroprotective microenvironment at the site of injury, which can help to reduce secondary damage, minimize neuronal death, and support the survival and integration of transplanted and endogenous neural cells.
5. Enhancement of Endogenous Repair Processes: Neural stem cells can stimulate endogenous repair processes within the brain, spinal cord, and nerves, including neurogenesis, synaptogenesis, and axonal sprouting. By enhancing these natural repair mechanisms, they contribute to tissue remodeling and functional recovery.
6.Formation of Supportive Microenvironment: Transplanted NSCs create a supportive microenvironment at the site of injury, facilitating tissue repair and remodeling. They interact with the surrounding cells and extracellular matrix, promoting cell-cell communication and tissue integration. Additionally, NSCs can modulate the biochemical and biophysical properties of the extracellular matrix, influencing cell behavior and promoting tissue regeneration[35-39].
1. Embryonic Stem Cells (ESCs): Derived from the inner cell mass of blastocysts, ESCs have the potential to differentiate into all cell types of the body, including neural cells.
2. Induced Pluripotent Stem Cells (iPSCs): Generated by reprogramming adult somatic cells, such as skin cells or blood cells, into a pluripotent state, iPSCs can be directed to differentiate into neural stem cells and progenitor stem cells.
3. Fetal Neural Stem Cells: Isolated from the developing fetal brain and spinal cord, fetal neural stem cells have the advantage of being already committed to a neural lineage and possessing high proliferative capacity.
4. Adult Neural Stem Cells: Found in specialized niches within the adult brain, such as the subventricular zone and the dentate gyrus of the hippocampus, adult neural stem cells can be isolated and expanded in vitro for transplantation purposes[40-44].
Mesenchymal stem cells (MSCs) are a type of adult stem cell that can be found in various tissues throughout the body, including bone marrow, adipose tissue, umbilical cord, and dental pulp. While MSCs are not inherently neural stem cells, they have been shown to possess neurogenic potential under certain conditions. When exposed to specific growth factors and signaling molecules in the appropriate microenvironment, MSCs can differentiate into cells with neural characteristics, including neurons, astrocytes, and oligodendrocytes. This phenomenon, known as transdifferentiation or lineage reprogramming, suggests that MSCs may serve as a potential source of neural stem cells for transplantation therapies aimed at repairing the brain, spinal cord, and nerves. Additionally, MSCs exert paracrine effects by secreting trophic factors and cytokines that promote neuroprotection, reduce inflammation, and enhance endogenous repair processes. While further research and clinical trials are needed to fully elucidate the neurogenic potential of MSCs and optimize their use in neural regeneration therapies, they represent a promising avenue for developing novel treatments for neurological disorders.
Each source of neural stem cells has its advantages and limitations in terms of availability, ethical considerations, immunogenicity, and differentiation potential, which need to be carefully considered when choosing an appropriate cell source for transplantation therapies[45-49].
Our Cellular Therapy and Neural Stem Cells operate through a sophisticated mechanism of action, intricately orchestrating neurogenic differentiation, neurotrophic factor secretion, immunomodulation, neuroprotective shielding, augmentation of endogenous repair, and niche modulation. Neural stem cells exhibit unparalleled plasticity, differentiating into diverse neural cell lineages upon transplantation, replenishing neuronal populations, and fortifying synaptic connections. Simultaneously, they unleash a potent arsenal of neurotrophic factors, fostering a regenerative milieu crucial for neuronal vitality and resilience. The immunomodulatory prowess of these cells calibrates the immune response, suppressing deleterious inflammation and nurturing neural repair. Serving as a neuroprotective shield, neural stem cells mitigate oxidative stress, neutralize excitotoxicity, and fortify neurons against adversity. Acting as catalysts for endogenous repair, they stimulate neurogenesis, synaptogenesis, and axonal regeneration, contributing to the rewiring of neural circuits. Finally, through niche modulation, transplanted neural stem cells sculpt a microenvironment conducive to cellular integration, structural reorganization, and functional recovery, encapsulating a transformative approach of Cellular Therapy and Stem Cells for Brain Spinal Cord and Neurodegenerative Diseases to neurological rejuvenation[50-54].
The key parameters serving as primary endpoints for evaluating the amelioration of patients post Cellular Therapy and Neural Stem Cell treatment for the listed conditions, along with standardized scoring methods where applicable, are as follows, listed in alphabetical order:
1. Alzheimer’s Disease: Cognitive function (assessed by Mini-Mental State Examination), memory retention, and functional independence.
2. Amyotrophic Lateral Sclerosis (ALS): Motor function (assessed by ALS Functional Rating Scale), muscle strength, and respiratory function.
3. Autism Spectrum Disorder (ASD): Social communication skills, behavioral patterns, and cognitive development.
4. Cerebellar Ataxia: Coordination, balance (assessed by Scale for the Assessment and Rating of Ataxia), and gait stability.
5. Cerebral Palsy (CP): Motor function, muscle tone, and mobility.
6. Dementia: Cognitive function (assessed by Mini-Mental State Examination), memory recall, and activities of daily living.
7. Epilepsy: Frequency and severity of seizures, medication dosage, and quality of life.
8. Hemorrhagic Stroke: Neurological deficits, motor function, and cognitive abilities.
9. Huntington’s Disease: Motor control, cognitive function (assessed by Unified Huntington’s Disease Rating Scale), and psychiatric symptoms.
10. Intellectual Disability: Adaptive behavior (assessed by Vineland Adaptive Behavior Scales), learning abilities, and social skills.
11. Ischemic Stroke: Neurological deficits, motor function, and cognitive abilities.
12. Multiple Sclerosis: Disease progression (assessed by Expanded Disability Status Scale), relapse rate, and neurological disability.
13. Multiple System Atrophy (MAS): Autonomic dysfunction, motor impairment (assessed by Unified Multiple System Atrophy Rating Scale), and quality of life.
14. Muscular Dystrophy (MD): Muscle strength (assessed by Manual Muscle Testing), mobility, and respiratory function.
15. Neuromuscular Disease (NMD): Muscle strength, motor function, and respiratory capacity.
16. Olfactory Disorder: Assessment of olfactory function (using tests such as Sniffin’ Sticks or University of Pennsylvania Smell Identification Test), improvement in odor detection and discrimination, and subjective reporting of olfactory perception
17. Parkinson’s Disease: Motor function (assessed by Unified Parkinson’s Disease Rating Scale), tremor severity, and activities of daily living.
18. Prion Disease: Disease progression (assessed by clinical symptoms such as cognitive decline, movement abnormalities, and psychiatric symptoms), quality of life, and neurological deficits.
19. Spinal cord injury: Motor and sensory function, bladder and bowel control, and mobility.
20. Spinal Muscular Atrophies (SMA): Muscle strength (assessed by Hammersmith Functional Motor Scale), motor function, and respiratory capacity.
21. Transient Ischemic Attack (TIA): Neurological deficits, motor function, and cognitive abilities.
22. Transverse Myelitis (TM): Motor and sensory function, bladder and bowel control, and quality of life.
23. Traumatic Brain Injury (TBI): Cognitive function (assessed by neuropsychological tests such as Trail Making Test or Glasgow Coma Scale), motor function, and activities of daily living. Additionally, imaging techniques like MRI or CT scans can be used to assess structural changes in the brain[55-62].
Our regimen of Cellular Therapy and Stem Cells for Brain Spinal Cord and Neurodegenerative Diseases entails a minimum duration of 10 to 14 days, comprising alternating infusions of Cellular Therapy enriched with Neural Growth Factors and intramuscular administration of Regenerative Exosomes with Peptide. In the optimal scenario for patients with varying severity levels of Brain, Spinal Cord, and Nerve diseases, our protocols prioritize the gradual regeneration of the body, particularly the nervous system. This approach of Cellular Therapy and Stem Cells for Brain Spinal Cord and Neurodegenerative Diseases contrasts with the practice of administering high doses of Cellular Therapy and Stem Cells in the shortest timeframe feasible, providing the body with the opportunity to regenerate at a more measured pace.
The graph shows improved NIHSS score in the treatment group (PEALut, green) after our special brain and spinal cord regeneration treatment protocols versus control group of patients with ischemic stroke (blue).
The graph shows a decreased number of patients with lumbosciatica in the treatment group (red) after our special brain and spinal cord regeneration treatment protocols versus control group (blue) at DrStemCellsThailand‘s Anti-Aging and Regenerative Medicine Center of Thailand.
Please refer to ongoing research and clinical trials around the world here!
The rationale behind the consistent recommendation of neurorehabilitation by our team of Regenerative physical therapists for individuals with Brain, Spinal Cord, and Nerve issues post-Cellular Therapy and Neural Stem Cell Treatment is multifaceted.
1. Functional Outcome Optimization: The primary goal is to enhance functional outcomes through targeted interventions aimed at maximizing neural recovery and adaptation.
2. Neuroplasticity Promotion: Facilitating neuroplasticity, the brain’s ability to reorganize and form new neural connections, is crucial for long-term neural recovery.
3. Independence Regain: Empowering individuals to regain independence is essential for improving quality of life and fostering a sense of autonomy.
4. Comprehensive Support: Providing comprehensive support addresses physical, cognitive, and emotional challenges associated with neurologic conditions, ensuring holistic care.
5. Task-Specific Exercises: Engaging in repetitive and task-specific exercises promotes neural rewiring and functional restoration, contributing to overall recovery.
6. Structured Interventions: Structured interventions, including physical, occupational, and speech therapy, play a vital role in addressing specific deficits and promoting recovery.
7. Holistic Approach: Embracing a holistic approach to neural rehabilitation ensures that all aspects of an individual’s well-being are addressed, leading to more robust and sustainable outcomes[63-70].
Contact us today for more information!
1.2 Amyotrophic Lateral Sclerosis (ALS)
1.3 Autism Spectrum Disorder (ASD)
1.6 Cerebrovascular Accident (CVA): Hemorrhagic Stroke, Ischemic Stroke
1.7 Dementia
1.8 Epilepsy
1.9 Huntington Disease (HD)
1.10 Intellectual Disability
1.12 Multiple System Atrophy (MAS)
1.14 Neuromuscular Disease (NMD)
1.15 Olfactory Disorders
1.17 Prion Disease
1.19 Spinal Muscular Atrophies (SMA)
1.21 Traumatic Brain Injury (TBI)
1.22 Brain tumors (e.g., glioblastoma)
1.23 Autism spectrum disorders (ASD)
1.24 Rett syndrome
1.25 Schizophrenia
1.26 Bipolar disorder
1.27 Major depressive disorder
1.28 Friedreich’s ataxia
1.29 Leukodystrophies
1.30 Ischemic brain injury
1.31 Retinal diseases (e.g., macular degeneration, retinitis pigmentosa)
1.32 Spinal muscular atrophy (SMA)
1.33 Leber’s hereditary optic neuropathy (LHON)
1.34 Ataxia telangiectasia
1.35 Neuronal ceroid lipofuscinosis (NCL)
1.36 Canavan disease
1.37 Sanfilippo syndrome
1.38 Metachromatic leukodystrophy (MLD)
1.39 Pelizaeus-Merzbacher disease
1.40 Neurodegenerative disorders associated with aging (e.g., age-related cognitive decline, age-related macular degeneration)
1.41 Motor Neuron Disease (MND)
Abbreviation
1. Mesenchymal Stem Cells (MSCs)
2. Hematopoietic Stem Cells (HSCs)
3. Induced Pluripotent Stem Cells (iPSCs)
4. Endothelial Progenitor Cells (EPCs)
5. Brain Progenitor Stem Cells (Brain-PSCs)
6. Umbilical Cord Stem Cells (UCSCs)
7. Adipose-Derived Stem Cells (ADSCs)
8. Dental Pulp Stem Cells (DPSCs)
9. Bone Marrow Mesenchymal Stem Cells (BMSCs)
1. Neuron Progenitor Stem Cells (Neuron-PSCs): Precursors that become different neuron types.
2. Astrocyte Progenitor Stem Cells (Astro-PSCs): Develop into structural and chemical support astrocytes.
3. Oligodendrocyte Progenitor Stem Cells (Oligo-PSCs): Transform into myelin-producing oligodendrocytes.
4. Microglial Progenitor Stem Cells (Micro-PSCs): Form immune-defense microglia cells.
5. Ependymal Progenitor Stem Cells (Ependy-PSCs): Give rise to cerebrospinal fluid-supporting ependymal cells.
Diseases associated with Brain, Spinal Cord and Peripheral Nerves |
Sources of Cellular Therapy&Neural Stem Cells |
Improvement Assessment by |
1.1.1. Early-Onset Alzheimer’s Disease (EOAD) 1.1.2. Late-Onset Alzheimer’s Disease (LOAD) 1.1.3. Familial Alzheimer’s Disease (FAD) 1.1.4. Sporadic Alzheimer’s Disease |
Fetal-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs, ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Cognitive Function: Evaluation of cognitive abilities using standardized tests such as the Mini-Mental State Examination (MMSE) or the Alzheimer’s Disease Assessment Scale-Cognitive Subscale (ADAS-Cog). 2. Activities of Daily Living (ADLs): Assessment of an individual’s ability to perform everyday tasks independently, including self-care, mobility, and household activities. 3. Neuropsychiatric Symptoms: Monitoring and evaluation of behavioral and psychological symptoms such as depression, agitation, and psychosis. 4. Disease Progression: Tracking changes in disease severity and progression over time through neuroimaging (e.g., MRI, PET scans) and biomarker analysis (e.g., cerebrospinal fluid biomarkers like Aβ and tau proteins). 5. Quality of Life: Assessing the overall quality of life and well-being of patients and caregivers using standardized scales and questionnaires. 6. Safety and Adverse Events: Monitoring and reporting any adverse events or side effects related to the cellular therapy or stem cell treatment, including infection, neurological complications, and immunological reactions. 7. Functional Independence: Evaluating the level of functional independence and autonomy in daily activities, mobility, and social interactions. 8. Neurological Examination: Conducting a comprehensive neurological assessment to detect changes in sensory, motor, and cognitive functions, as well as reflexes and coordination. 9. Global Clinical Impression: Obtaining overall clinical impressions and judgments from healthcare professionals regarding changes in symptoms, disease course, and treatment response. 10. Biomarker Analysis: Utilizing specific biomarkers related to AD pathology, such as amyloid-beta (Aβ) and tau protein levels, to assess disease progression and treatment effects at a molecular level.
Consult with Our Team of Experts Now!
|
1.2 Amyotrophic Lateral Sclerosis (ALS) 1.2.1. Sporadic ALS (sALS) 1.2.2. Familial ALS (fALS) 1.2.3. Bulbar ALS 1.2.4. Limb-Onset ALS 1.2.5. Progressive Muscular Atrophy (PMA) 1.2.6. Primary Lateral Sclerosis (PLS) |
Fetal-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Motor Function: Assessment of motor abilities using standardized scales such as the ALS Functional Rating Scale-Revised (ALSFRS-R) to measure muscle strength, speech, swallowing, and respiratory function. 2. Disease Progression: Monitoring and evaluation of disease progression over time through measures such as forced vital capacity (FVC), muscle strength testing, and electromyography (EMG) studies. 3. Respiratory Function: Evaluation of respiratory muscle strength, pulmonary function, and respiratory symptoms using spirometry, peak cough flow, and respiratory questionnaires. 4. Quality of Life: Assessing the overall quality of life, well-being, and emotional health of patients and caregivers using standardized quality-of-life scales and questionnaires. 5. Functional Independence: Evaluating the level of functional independence in daily activities, mobility, communication, and self-care tasks. 6. Survival Analysis: Tracking survival rates and time to disease progression or milestone events such as tracheostomy or initiation of invasive ventilation. 7. Neurological Examination: Conducting a comprehensive neurological assessment to detect changes in sensory, motor, and bulbar functions, reflexes, and coordination. 8. Bulbar Function: Specifically assessing bulbar function, including speech, swallowing, and facial muscle strength, which are often affected in bulbar-onset ALS subtypes. 9. Cognitive Function: Screening for cognitive and behavioral changes using tests such as the Edinburgh Cognitive and Behavioral ALS Screen (ECAS) or Frontal Assessment Battery (FAB). 10. Adverse Events Monitoring: Monitoring and reporting any adverse events, complications, or side effects related to cellular therapy or stem cell treatments, including infections, neurological complications, and immune reactions.
Consult with Our Team of Experts Now!
|
1.3 Autism Spectrum Disorder (ASD) 1.3.2. Asperger’s Syndrome 1.3.3. Childhood Disintegrative Disorder (CDD) 1.3.4. Pervasive Developmental Disorder-Not Otherwise Specified (PDD-NOS) 1.3.5. Rett Syndrome 1.3.6. Fragile X Syndrome |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Behavioral Symptoms: Assessment of core ASD symptoms, including social communication deficits, repetitive behaviors, restricted interests, and sensory sensitivities, using standardized scales such as the Autism Diagnostic Observation Schedule (ADOS) and Autism Diagnostic Interview-Revised (ADI-R). 2. Communication Skills: Evaluation of language development, speech articulation, expressive and receptive language abilities, and pragmatic language skills. 3. Social Interaction: Measurement of social skills, social reciprocity, peer interactions, and understanding of social cues and nonverbal communication. 4. Adaptive Functioning: Assessment of adaptive skills related to daily living activities, self-care, functional independence, and community integration. 5. Cognitive Abilities: Screening for cognitive strengths and weaknesses, intellectual functioning, executive function skills, and cognitive flexibility. 6. Emotional Regulation: Evaluation of emotional regulation, emotional expression, coping strategies, and responses to stress and anxiety. 7. Sensory Processing: Monitoring sensory processing difficulties, sensory seeking or aversion behaviors, and sensory integration challenges. 8. Quality of Life: Assessing the overall quality of life, well-being, and satisfaction of individuals with ASD and their families using standardized quality-of-life measures and questionnaires. 9. Family Functioning: Evaluation of family dynamics, caregiver stress, parental coping strategies, and family support networks. 10. Safety and Adverse Events: Monitoring and reporting any adverse events, side effects, or complications related to cellular therapy or stem cell treatments, including behavioral changes, medical issues, and neurological reactions.
Consult with Our Team of Experts Now!
|
1.4 Cerebellar Ataxia 1.4.2. Friedreich’s Ataxia 1.4.3. Ataxia Telangiectasia 1.4.4. Multiple System Atrophy with Cerebellar Ataxia (MSA-C) 1.4.5. Cerebellar Atrophy (Non-specific) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Motor Coordination: Assessment of motor coordination and balance using standardized scales such as the Scale for the Assessment and Rating of Ataxia (SARA) and the International Cooperative Ataxia Rating Scale (ICARS). 2. Gait Analysis: Evaluation of walking patterns, gait stability, step length, and base of support during walking tasks. 3. Fine Motor Skills: Measurement of fine motor abilities, hand dexterity, finger tapping speed, and precision tasks. 4. Speech and Swallowing: Assessment of speech articulation, vocal quality, dysarthria severity, and swallowing difficulties. 5. Eye Movement Control: Evaluation of eye movement coordination, saccades, pursuit movements, and nystagmus. 6. Activities of Daily Living (ADLs): Monitoring the ability to perform daily living tasks independently, including self-care, dressing, feeding, and mobility. 7. Quality of Life: Assessing the overall quality of life, functional independence, participation in social activities, and emotional well-being using standardized quality-of-life measures and questionnaires. 8. Neurological Examination: Conducting a comprehensive neurological assessment to detect changes in sensory, motor, and cerebellar functions, reflexes, and coordination. 9. Disease Progression: Tracking changes in disease severity, functional decline, and progression of ataxia symptoms over time through clinical evaluations and neuroimaging studies. 10. Safety and Adverse Events: Monitoring and reporting any adverse events, side effects, or complications related to cellular therapy or stem cell treatments, including neurological symptoms, infections, and immune reactions. Consult with Our Team of Experts Now!
|
1.5 Cerebral Palsy (CP) 1.5.1. Spastic Cerebral Palsy 1.5.2. Dyskinetic Cerebral Palsy 1.5.3. Ataxic Cerebral Palsy 1.5.4. Mixed Cerebral Palsy |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Motor Function: Assessment of motor function and mobility using standardized scales such as the Gross Motor Function Classification System (GMFCS) and the Manual Ability Classification System (MACS). 2. Muscle Tone: Evaluation of muscle tone, spasticity, rigidity, and dystonia using clinical assessments and tools like the Modified Ashworth Scale (MAS) and the Tardieu Scale. 3. Functional Independence: Measurement of functional independence in daily activities, self-care tasks, mobility, and participation in home, school, and community environments. 4. Communication Skills: Assessment of communication abilities, speech intelligibility, language development, augmentative communication use, and social communication skills. 5. Activities of Daily Living (ADLs): Monitoring the ability to perform daily living tasks independently, including dressing, feeding, grooming, toileting, and mobility. 6. Quality of Life: Assessing the overall quality of life, well-being, social integration, and participation in recreational and leisure activities using standardized quality-of-life measures and questionnaires. 7. Cognitive Function: Screening for cognitive abilities, intellectual functioning, executive function skills, attention, memory, and learning abilities. 8. Pain Assessment: Evaluation of pain levels, discomfort, and musculoskeletal issues related to CP using pain scales, patient reports, and caregiver observations. 9. Neurological Examination: Conducting a comprehensive neurological assessment to detect changes in sensory, motor, and cognitive functions, reflexes, and coordination. 10. Safety and Adverse Events: Monitoring and reporting any adverse events, side effects, or complications related to cellular therapy or stem cell treatments, including infections, neurological symptoms, and immune reactions. Consult with Our Team of Experts Now!
|
1.6 Cerebrovascular Accident (CVA) 1.6.1. Ischemic Stroke 1.6.2. Hemorrhagic Stroke 1.6.3. Transient Ischemic Attack (TIA) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Neurological Function: Assessment of neurological deficits, motor function, sensory function, speech and language abilities, and cognitive function using standardized scales such as the National Institutes of Health Stroke Scale (NIHSS) and the Modified Rankin Scale (mRS). 2. Functional Independence: Evaluation of functional independence in activities of daily living (ADLs), mobility, self-care tasks, and instrumental activities of daily living (IADLs) using tools like the Barthel Index and the Functional Independence Measure (FIM). 3. Motor Recovery: Measurement of motor recovery, motor control, muscle strength, range of motion, spasticity, and coordination through clinical evaluations and assessments like the Fugl-Meyer Assessment (FMA) and the Action Research Arm Test (ARAT). 4. Cognitive Assessment: Screening for cognitive impairments, attention deficits, memory problems, executive function deficits, and language difficulties using neuropsychological tests and assessments such as the Montreal Cognitive Assessment (MoCA) and the Mini-Mental State Examination (MMSE). 5. Quality of Life: Assessing the overall quality of life, emotional well-being, social participation, and satisfaction with life using standardized quality-of-life measures and questionnaires. 6. Activities of Daily Living (ADLs): Monitoring the ability to perform activities of daily living independently, including dressing, grooming, toileting, feeding, and household tasks. 7. Speech and Language Function: Evaluation of speech articulation, fluency, voice quality, language comprehension, expression, and communication skills using speech-language assessments and tools. 8. Swallowing Function: Assessment of swallowing function, dysphagia severity, risk of aspiration, and oral motor control using clinical swallowing evaluations and swallowing assessments. 9. Safety and Adverse Events: Monitoring and reporting any adverse events, complications, or side effects related to cellular therapy or stem cell treatments, including infections, neurological complications, and immune reactions. Consult with Our Team of Experts Now!
|
1.7 Dementia 1.7.1. Alzheimer’s Disease (AD) 1.7.2. Vascular Dementia 1.7.3. Lewy Body Dementia (LBD) 1.7.4. Frontotemporal Dementia (FTD) 1.7.5. Mixed Dementia 1.7.6. Parkinson’s Disease Dementia (PDD) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs, MSCs-Exos Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Cognitive Function: Evaluation of cognitive abilities including memory, attention, executive function, language, and visuospatial skills using standardized tests such as the Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment (MoCA), and Alzheimer’s Disease Assessment Scale-Cognitive Subscale (ADAS-Cog). 2. Functional Independence: Assessment of functional independence in daily living activities such as self-care, mobility, household tasks, and financial management using scales like the Activities of Daily Living (ADLs) and Instrumental Activities of Daily Living (IADLs) scales. 3. Behavioral and Psychological Symptoms: Monitoring and evaluation of behavioral and psychological symptoms such as agitation, aggression, apathy, depression, hallucinations, and delusions using standardized scales like the Neuropsychiatric Inventory (NPI) and the Cornell Scale for Depression in Dementia (CSDD). 4. Quality of Life: Assessing the overall quality of life, well-being, and satisfaction of patients and caregivers using standardized quality-of-life measures and questionnaires. 5. Neuropsychiatric Symptoms: Evaluation of neuropsychiatric symptoms including sleep disturbances, anxiety, irritability, and disinhibition using scales like the Behavioral Pathology in Alzheimer’s Disease Rating Scale (BEHAVE-AD) and the Pittsburgh Agitation Scale (PAS). 6. Disease Progression: Tracking changes in disease severity, cognitive decline, functional abilities, and behavioral symptoms over time through clinical assessments and neuroimaging studies. 7. Safety and Adverse Events: Monitoring and reporting any adverse events, complications, or side effects related to cellular therapy or stem cell treatments, including infections, neurological complications, and immune reactions. 8. Neurological Examination: Conducting a comprehensive neurological assessment to detect changes in sensory, motor, and cognitive functions, reflexes, and coordination. 9. Caregiver Burden: Assessing the impact of dementia on caregivers’ emotional well-being, physical health, social support, and caregiving responsibilities using caregiver burden scales and questionnaires. Consult with Our Team of Experts Now!
|
1.8 Epilepsy 1.8.1. Focal (Partial) Epilepsy 1.8.2. Generalized Epilepsy 1.8.3. Absence Seizures 1.8.4. Tonic-Clonic Seizures 1.8.5. Temporal Lobe Epilepsy (TLE) 1.8.6. Frontal Lobe Epilepsy 1.8.7. Occipital Lobe Epilepsy 1.8.8. Juvenile Myoclonic Epilepsy (JME) 1.8.9. Lennox-Gastaut Syndrome (LGS) 1.8.10. Dravet Syndrome |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Seizure Frequency: Monitoring and assessment of seizure frequency, type, duration, and severity using seizure diaries, caregiver reports, and seizure frequency scales. 2. Seizure Control: Evaluation of seizure control and response to treatment, including the number of seizure-free periods, reduction in seizure frequency, and seizure severity. 3. Antiepileptic Medication Use: Assessment of antiepileptic medication usage, dosage adjustments, adherence to treatment, and changes in medication regimens post-cellular therapy. 4. Quality of Life: Measuring the impact of epilepsy on quality of life, emotional well-being, social interactions, employment, education, and activities of daily living using standardized quality-of-life scales and questionnaires. 5. Neurological Examination: Conducting a comprehensive neurological assessment to detect changes in sensory, motor, and cognitive functions, reflexes, coordination, and neurological signs related to epilepsy. 6. Cognitive Function: Screening for cognitive impairments, memory deficits, attention problems, executive function deficits, and language difficulties using neuropsychological tests and assessments. 7. Adverse Events: Monitoring and reporting any adverse events, complications, or side effects related to cellular therapy or stem cell treatments, including neurological symptoms, infections, and immune reactions. 8. Functional Independence: Evaluating the level of functional independence in daily activities, mobility, self-care tasks, and participation in school, work, or community settings. 9. Behavioral and Psychological Symptoms: Assessment of behavioral and psychological symptoms such as mood changes, anxiety, depression, aggression, and impulsivity using standardized scales and questionnaires. Consult with Our Team of Experts Now!
|
1.9.1. Huntington’s Disease (Classic) 1.9.2. Juvenile Huntington’s Disease 1.9.3. Late-Onset Huntington’s Disease 1.9.4. Presymptomatic Huntington’s Disease |
Adult&Fetal-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs ,MSCs-Exos Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Motor Function: Evaluation of motor function, including chorea (involuntary movements), dystonia (muscle contractions), bradykinesia (slowness of movement), gait abnormalities, and motor coordination using scales such as the Unified Huntington’s Disease Rating Scale (UHDRS) and the Total Motor Score (TMS). 2. Cognitive Function: Assessment of cognitive abilities, including memory, attention, executive function, language, and visuospatial skills using neuropsychological tests such as the Symbol Digit Modalities Test (SDMT) and the Stroop Test. 3. Functional Independence: Measurement of functional independence in activities of daily living (ADLs), instrumental activities of daily living (IADLs), self-care tasks, and mobility using scales like the Functional Independence Measure (FIM) and the Barthel Index. 4. Psychiatric Symptoms: Evaluation of psychiatric symptoms, including depression, anxiety, irritability, apathy, and psychosis using scales like the Beck Depression Inventory (BDI) and the Neuropsychiatric Inventory (NPI). 5. Quality of Life: Assessing the overall quality of life, emotional well-being, social integration, and satisfaction with life using standardized quality-of-life measures and questionnaires. 6. Neurological Examination: Conducting a comprehensive neurological assessment to detect changes in sensory, motor, and cognitive functions, reflexes, coordination, and neurological signs related to Huntington’s Disease. 7. Safety and Adverse Events: Monitoring and reporting any adverse events, complications, or side effects related to cellular therapy or stem cell treatments, including infections, neurological symptoms, and immune reactions. 8. Genetic Biomarkers: Utilizing genetic biomarkers and molecular assays to track disease progression, identify therapeutic targets, and assess treatment response at a molecular level. Consult with Our Team of Experts Now!
|
1. Severity: – Mild ID: IQ scores between 50-70, needing some support. – Moderate ID: IQ scores between 35-49, requiring more significant support. – Severe ID: IQ scores between 20-34, needing extensive support. – Profound ID: IQ scores below 20, requiring constant care. 2. Associated Features: – Developmental Delay: Common in individuals with ID. – Behavioral and Emotional Challenges: Some may experience behavior or emotional regulation difficulties. – Co-occurring Conditions: Often seen alongside other conditions like autism, ADHD, epilepsy, or physical disabilities. |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Cognitive Function: Assessment of cognitive abilities, including intellectual functioning, adaptive behavior skills, memory, attention, problem-solving, and executive function, using standardized tests such as the Wechsler Intelligence Scale for Children (WISC) or the Vineland Adaptive Behavior Scales (VABS). 2. Adaptive Behavior: Evaluation of adaptive behavior skills in daily living activities, communication, socialization, self-care, home living, community participation, and functional independence using scales like the Adaptive Behavior Assessment System (ABAS) or the Scales of Independent Behavior-Revised (SIB-R). 3. Communication Skills: Measurement of communication abilities, including expressive and receptive language skills, speech articulation, pragmatic language use, and alternative communication methods. 4. Motor Skills: Assessment of motor skills, coordination, fine motor abilities, gross motor abilities, and physical functioning. 5. Social and Emotional Functioning: Evaluation of social skills, emotional regulation, behavior management, social interactions, peer relationships, and emotional well-being using standardized scales and behavioral assessments. 6. Quality of Life: Assessing the overall quality of life, well-being, satisfaction, and participation in meaningful activities using standardized quality-of-life measures and questionnaires. 7. Functional Independence: Monitoring the level of functional independence in daily activities, self-care tasks, vocational skills, and community integration. 8. Safety and Adverse Events: Monitoring and reporting any adverse events, complications, or side effects related to cellular therapy or stem cell treatments, including behavioral changes, medical issues, and neurological symptoms. Consult with Our Team of Experts Now!
|
1.11.1. Relapsing-Remitting Multiple Sclerosis (RRMS) 1.11.2. Secondary Progressive Multiple Sclerosis (SPMS) 1.11.3. Primary Progressive Multiple Sclerosis (PPMS) 1.11.4. Clinically Isolated Syndrome (CIS) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Neurological Function: Assessment of neurological symptoms, disability progression, motor function, sensory function, coordination, and ambulation using scales such as the Expanded Disability Status Scale (EDSS) and the Multiple Sclerosis Functional Composite (MSFC). 2. Disease Activity: Monitoring disease activity, relapse rates, MRI lesions, gadolinium-enhancing lesions, T2 lesion volume, and brain atrophy using neuroimaging techniques and MRI scans. 3. Cognitive Function: Evaluation of cognitive abilities, including memory, attention, processing speed, executive function, verbal fluency, and visuospatial skills using neuropsychological tests such as the Symbol Digit Modalities Test (SDMT) and the Paced Auditory Serial Addition Test (PASAT). 4. Fatigue Assessment: Measurement of fatigue severity, impact on daily activities, and fatigue-related impairment using fatigue scales and questionnaires. 5. Quality of Life: Assessing the overall quality of life, emotional well-being, social functioning, and satisfaction with life using standardized quality-of-life measures and questionnaires specific to MS. 6. Ambulatory Function: Evaluation of ambulatory function, walking speed, endurance, balance, and mobility using assessments like the Timed 25-Foot Walk (T25FW) and the 6-Minute Walk Test (6MWT). 7. Visual Function: Monitoring visual acuity, contrast sensitivity, color vision, visual field defects, and ocular motor function using ophthalmologic examinations and visual function tests. 8. Bladder and Bowel Function: Assessment of bladder and bowel dysfunction, including frequency, urgency, incontinence, and bowel habits using bladder diaries, bowel function questionnaires, and urodynamic studies. 9. Mood and Mental Health: Screening for mood disorders, anxiety, depression, emotional well-being, and mental health concerns using standardized assessments and psychiatric evaluations. 10. Safety and Adverse Events: Monitoring and reporting any adverse events, complications, or side effects related to cellular therapy or stem cell treatments, including infections, neurological symptoms, and immune reactions. Consult with Our Team of Experts Now!
|
1.12 Multiple System Atrophy (MAS): 1.12.1. MSA with Predominant Parkinsonism (MSA-P) 1.12.2. MSA with Predominant Cerebellar Ataxia (MSA-C) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs Doses : A three-stage dose escalation scheme (low-dose, 3.0 × 105 cells/kg; medium-dose, 6.0 × 105 cells/kg; high-dose, 9.0 × 105 cells/kg) was applied to determine the maximum tolerated dose of intra-arterial administration of BM-MSC |
1. Motor Function: Assessment of motor symptoms, including parkinsonism, ataxia, rigidity, tremor, bradykinesia, gait disturbances, and coordination using standardized scales such as the Unified Multiple System Atrophy Rating Scale (UMSARS) and the International Cooperative Ataxia Rating Scale (ICARS). 2. Autonomic Function: Evaluation of autonomic dysfunction, including orthostatic hypotension, urinary dysfunction (e.g., urinary urgency, incontinence), constipation, and dysphagia using clinical assessments and autonomic function tests. 3. Cognitive Function: Screening for cognitive impairments, memory deficits, executive function deficits, attention problems, and language difficulties using neuropsychological tests and assessments. 4. Quality of Life: Assessing the overall quality of life, emotional well-being, social functioning, and satisfaction with life using standardized quality-of-life measures and questionnaires specific to MSA. 5. Functional Independence: Measurement of functional independence in daily activities, mobility, self-care tasks, and instrumental activities of daily living (IADLs) using scales like the Activities of Daily Living (ADLs) and Instrumental Activities of Daily Living (IADLs) scales. 6. Safety and Adverse Events: Monitoring and reporting any adverse events, complications, or side effects related to cellular therapy or stem cell treatments, including infections, neurological symptoms, and immune reactions. 7. Disease Progression: Tracking changes in disease severity, motor and autonomic symptoms, disability progression, and overall functional decline over time through clinical evaluations and neuroimaging studies. Consult with Our Team of Experts Now!
|
1.13 Muscular Dystrophy 1.13.1 Duchenne Muscular Dystrophy (DMD) 1.13.2 Becker Muscular Dystrophy (BMD) 1.13.3 Myotonic Dystrophy 1.13.4 Facioscapulohumeral Muscular Dystrophy (FSHD) 1.13.5 Limb-Girdle Muscular Dystrophy (LGMD) 1.13.6 Congenital Muscular Dystrophy (CMD) 1.13.7 Oculopharyngeal Muscular Dystrophy (OPMD) 1.13.8 Emery-Dreifuss Muscular Dystrophy (EDMD) 1.13.9 Distal Muscular Dystrophy (DD) 1.13.10 Ullrich Congenital Muscular Dystrophy (UCMD) 1.13.11 Bethlem Myopathy 1.13.12 Miyoshi Myopathy 1.13.13 Distal Myopathies (DM) 1.13.14 Emery-Dreifuss-like Phenotype 1.13.15 Other Unspecified Muscular Dystrophies |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Improvement in muscle strength and function: This can be assessed using standardized tests such as manual muscle testing (e.g., Medical Research Council (MRC) scale), handheld dynamometry, or functional assessments like the six-minute walk test (6MWT) and timed function tests (e.g., time to rise from the floor, time to climb stairs). 2. Reduction in muscle degeneration and wasting: Evaluation may involve muscle imaging techniques such as MRI or ultrasound to assess changes in muscle size, composition, and fat infiltration. Muscle biopsy may also be performed to analyze histological changes in muscle tissue. 3. Increase in muscle mass: Measurement of muscle mass can be done through imaging techniques like dual-energy X-ray absorptiometry (DEXA) or MRI, which provide information about muscle volume and composition changes. 4. Improvement in mobility and ability to perform daily activities: Functional assessments such as the North Star Ambulatory Assessment (NSAA), Motor Function Measure (MFM), and Activities of Daily Living (ADL) scales can be used to evaluate improvements in mobility, motor skills, and ability to perform daily tasks. 5. Reduction in pain and discomfort associated with MD: Pain scales (e.g., visual analog scale (VAS), Numerical Rating Scale (NRS)) can be used to quantify pain levels and assess changes post-treatment. 6. Improvement in respiratory function and breathing capacity: Pulmonary function tests (e.g., spirometry, maximal inspiratory and expiratory pressures) can assess respiratory muscle strength and lung function, while sleep studies (polysomnography) can evaluate sleep-disordered breathing and respiratory function during sleep. 7. Increase in overall quality of life and well-being: Quality of life assessments using standardized questionnaires such as the SF-36, PedsQL (Pediatric Quality of Life Inventory), or MD-specific quality of life scales can measure improvements in physical, emotional, and social well-being. Consult with Our Team of Experts Now!
|
1.14 Neuromuscular Disease (NMD) 1.14.1 Duchenne Muscular Dystrophy (DMD) 1.14.2 Becker Muscular Dystrophy (BMD) 1.14.3 Myotonic Dystrophy 1.14.4 Facioscapulohumeral Muscular Dystrophy (FSHD) 1.14.5 Limb-Girdle Muscular Dystrophy (LGMD) 1.14.6 Spinal Muscular Atrophy (SMA) 1.14.7 Charcot-Marie-Tooth Disease (CMT) 1.14.8 Amyotrophic Lateral Sclerosis (ALS) 1.14.9 Myasthenia Gravis (MG) 1.14.10 Hereditary Neuropathies 1.14.11 Inflammatory Myopathies : 1.14.11.1 Dermatomyositis 1.14.11.2 Polymyositis 1.14.11.3 Inclusion Body Myositis 1.14.12 Metabolic Myopathies 1.14.13 Motor Neuron Diseases 1.14.14 Muscular Dystrophies (other than Duchenne and Becker) 1.14.15 Peripheral Neuropathies 1.14.16 Neuromuscular Junction Disorders 1.14.17 Mitochondrial Myopathies |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Improvement in muscle strength and function: Assessment can involve standardized tests such as manual muscle testing, handheld dynamometry, or functional tests like the 6-minute walk test (6MWT), timed function tests, and muscle strength grading scales (e.g., Medical Research Council (MRC) scale). 2. Reduction in muscle degeneration and wasting: Evaluation may include imaging techniques such as MRI or ultrasound to assess changes in muscle size, composition, and fat infiltration. Muscle biopsy can provide histological information on muscle tissue integrity and degeneration. 3. Increase in muscle mass: Measurement of muscle mass can be conducted using imaging modalities like DEXA or MRI, which provide insights into changes in muscle volume and composition. 4. Improvement in mobility and ability to perform daily activities: Functional assessments such as the North Star Ambulatory Assessment (NSAA), Motor Function Measure (MFM), and Activities of Daily Living (ADL) scales can evaluate improvements in mobility, motor skills, and the ability to perform daily tasks. 5. Reduction in pain and discomfort associated with NMD: Pain scales (e.g., visual analog scale (VAS), Numerical Rating Scale (NRS)) can quantify pain levels and assess changes post-treatment. 6. Improvement in nerve conduction and electromyography (EMG) findings: Electrophysiological tests, including nerve conduction studies and EMG, can assess changes in nerve function, muscle activation patterns, and motor unit recruitment. 7. Increase in overall quality of life and well-being: Quality of life assessments using standardized questionnaires such as the SF-36, PedsQL, or disease-specific quality of life scales can measure improvements in physical, emotional, and social well-being. 8. Improvement in respiratory function and breathing capacity: Pulmonary function tests (e.g., spirometry, maximal inspiratory and expiratory pressures) can assess respiratory muscle strength and lung function, while sleep studies (polysomnography) can evaluate sleep-disordered breathing and respiratory function during sleep. 9. Reduction in disease progression or stabilization of symptoms: Longitudinal assessment of disease progression through clinical measures, functional tests, and imaging can indicate whether the disease course has been altered or stabilized post-treatment. Consult with Our Team of Experts Now!
|
1.15 Olfactory Disorders
1.15.1 Post-viral Olfactory Dysfunction (PVOD) 1.15.2 Idiopathic Olfactory Loss 1.15.3 Sinonasal Olfactory Disorders (e.g., chronic rhinosinusitis-associated olfactory loss) 1.15.4 Neurodegenerative Olfactory Disorders (e.g., Parkinson’s disease-associated olfactory dysfunction) 1.15.5 Traumatic Olfactory Disorders (e.g., head injury-induced olfactory loss) 1.15.6 Toxic Olfactory Disorders (e.g., chemical exposure-induced olfactory dysfunction) 1.15.7 Age-related Olfactory Dysfunction (presbyosmia) 1.15.8 Olfactory Disorders Associated with Systemic Diseases (e.g., diabetes mellitus-related olfactory dysfunction) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Improvement in olfactory function: – Objective measures such as olfactory testing using standardized smell identification tests like the University of Pennsylvania Smell Identification Test (UPSIT) or Sniffin’ Sticks test. – Subjective assessments through patient-reported outcomes and self-assessment questionnaires regarding changes in smell perception and discrimination. 2. Restoration of olfactory neuroepithelium: – Evaluation of nasal mucosa integrity and regeneration through nasal endoscopy or biopsy to assess changes in the olfactory epithelium structure and function. 3. Reduction in olfactory dysfunction-related symptoms: – Assessment of symptoms such as anosmia (loss of smell), hyposmia (reduced sense of smell), parosmia (distorted sense of smell), and phantosmia (perception of odor in the absence of external stimuli) through patient-reported outcomes and symptom severity scales. 4. Improvement in quality of life: – Utilization of validated quality of life measures specific to olfactory dysfunction, such as the Questionnaire of Olfactory Disorders-Negative Statements (QOD-NS) or the Olfactory Disorders Quality of Life Scale (ODQoL). 5. Restoration of taste function (if affected): – Evaluation of taste perception and discrimination through taste tests and self-reported changes in taste sensation. Consult with Our Team of Experts Now!
|
1.16.1 Idiopathic Parkinson’s Disease (classic or typical PD) 1.16.2 Atypical Parkinsonian Syndromes (e.g., Multiple System Atrophy (MSA), Progressive Supranuclear Palsy (PSP), Corticobasal Degeneration (CBD)) 1.16.3 Parkinsonism-plus Syndromes 1.16.4 Drug-induced Parkinsonism 1.16.5 Vascular Parkinsonism 1.16.6 Parkinsonism due to other causes (e.g., traumatic brain injury, toxins) |
Fetal&Adult-NSCs,BM‐MSCs,hUCB‐MSCs,DPSCs,ESCs, iPSCs, MSCs-Exos Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Improvement in motor symptoms: – Evaluation using standardized rating scales such as the Unified Parkinson’s Disease Rating Scale (UPDRS) or the Movement Disorder Society Unified Parkinson’s Disease Rating Scale (MDS-UPDRS), focusing on measures of tremor, rigidity, bradykinesia, and postural instability. 2. Reduction in non-motor symptoms: – Assessment of non-motor symptoms such as cognitive impairment (using scales like the Montreal Cognitive Assessment), mood disturbances (e.g., depression, anxiety), sleep disturbances, autonomic dysfunction, and sensory symptoms. 3. Improvement in activities of daily living (ADLs): – Measurement of functional abilities and independence in daily activities using scales such as the Schwab and England Activities of Daily Living Scale (SE-ADL), Parkinson’s Disease Activities of Daily Living Scale (PADLS), or the Parkinson’s Disease Questionnaire (PDQ-39). 4. Increase in quality of life: – Utilization of validated quality of life measures specific to Parkinson’s disease, such as the Parkinson’s Disease Quality of Life Questionnaire (PDQ-8) or the Parkinson’s Disease Questionnaire (PDQ-39). 5. Long-term disease modification: – Evaluation of disease progression through longitudinal assessment of motor and non-motor symptoms, functional decline, and changes in medication requirements over time. 6. Reduction in medication-related complications: – Assessment of medication-induced side effects such as dyskinesias, motor fluctuations (e.g., wearing-off, on-off phenomena), and psychiatric complications (e.g., impulse control disorders). 7. Improvement in neuroimaging markers: – Utilization of neuroimaging techniques such as MRI, PET, or SPECT to assess changes in brain structure, function, and connectivity following treatment. Consult with Our Team of Experts Now!
|
1.17 Prion Disease 1.17.1 Creutzfeldt-Jakob Disease (CJD) 1.17.2 Variant Creutzfeldt-Jakob Disease (vCJD) 1.17.3 Gerstmann-Sträussler-Scheinker Syndrome (GSS) 1.17.4 Fatal Familial Insomnia (FFI) 1.17.5 Kuru 1.17.6 Variably Protease-Sensitive Prionopathy (VPSPr) 1.17.7 Iatrogenic Creutzfeldt-Jakob Disease (iCJD) 1.17.8 Sporadic Fatal Insomnia (SFI) 1.17.9 Familial Creutzfeldt-Jakob Disease (fCJD) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Stabilization or improvement in cognitive function: – Assessment using neuropsychological tests to evaluate changes in memory, attention, language, executive function, and visuospatial abilities. 2. Reduction in disease progression: – Evaluation of clinical symptoms and signs associated with prion disease progression, including changes in gait, coordination, motor function, and neurological deficits. 3. Preservation of activities of daily living (ADLs): – Measurement of functional independence and ability to perform daily tasks such as self-care, mobility, and communication. 4. Improvement in quality of life: – Utilization of standardized quality of life measures to assess changes in physical, emotional, and social well-being. 5. Reduction in disease-associated complications: – Assessment of symptoms such as myoclonus, seizures, psychiatric disturbances, and autonomic dysfunction. 6. Long-term survival and disease-free intervals: – Evaluation of overall survival rates and disease-free intervals following treatment with cellular therapy and stem cells. Consult with Our Team of Experts Now!
|
1.18.1 Complete Spinal Cord Injury 1.18.2 Incomplete Spinal Cord Injury (e.g., Brown-Sequard Syndrome, Central Cord Syndrome, Anterior Cord Syndrome) 1.18.3 Tetraplegia (Quadriplegia) 1.18.4 Paraplegia 1.18.5 Cervical Spinal Cord Injury 1.18.6 Thoracic Spinal Cord Injury 1.18.7 Lumbar Spinal Cord Injury 1.18.8 Sacral Spinal Cord Injury 1.18.9 Traumatic Spinal Cord Injury 1.18.10 Non-traumatic Spinal Cord Injury (e.g., due to tumors, infections, ischemia) |
AF-MSCs,Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Improvement in motor function: – Assessment using standardized motor scales such as the International Standards for Neurological Classification of Spinal Cord Injury (ISNCSCI) or the ASIA Impairment Scale (AIS) to evaluate changes in muscle strength, sensation, and motor function. 2. Enhancement of sensory function: – Evaluation of sensory modalities (light touch, pinprick, proprioception) using standardized sensory scales to assess improvements in sensory perception and discrimination. 3. Restoration of autonomic function: – Assessment of autonomic dysreflexia, bladder and bowel function, sexual function, and cardiovascular function to determine improvements in autonomic regulation post-treatment. 4. Reduction in spasticity and neuropathic pain: – Measurement of spasticity using scales such as the Modified Ashworth Scale (MAS) and assessment of neuropathic pain using pain scales to quantify changes in spasticity and pain levels. 5. Improvement in activities of daily living (ADLs): – Evaluation of functional independence and ability to perform daily tasks such as self-care, mobility, transfers, and wheelchair use. 6. Restoration of bladder and bowel function: – Assessment of bladder and bowel continence, voiding patterns, and bowel movements to determine improvements in bladder and bowel function post-treatment. 7. Increase in quality of life: – Utilization of validated quality of life measures specific to spinal cord injury, such as the Spinal Cord Independence Measure (SCIM), the Craig Handicap Assessment and Reporting Technique (CHART), or the World Health Organization Quality of Life Scale (WHOQOL-BREF). Consult with Our Team of Experts Now!
|
1.19 Spinal Muscular Atrophies (SMA) 1.19.1 Spinal Muscular Atrophy Type 1 (SMA Type 1 or Werdnig-Hoffmann Disease) 1.19.2 Spinal Muscular Atrophy Type 2 (SMA Type 2) 1.19.3 Spinal Muscular Atrophy Type 3 (SMA Type 3 or Kugelberg-Welander Disease) 1.19.4 Spinal Muscular Atrophy Type 4 (Adult-onset SMA) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Improvement in motor function: – Assessment using standardized motor scales such as the Hammersmith Functional Motor Scale-Expanded (HFMSE) or the Revised Upper Limb Module (RULM) to evaluate changes in motor skills, muscle strength, and functional abilities. 2. Preservation or increase in muscle strength: – Evaluation of muscle strength using manual muscle testing (e.g., Medical Research Council (MRC) scale), handheld dynamometry, or functional tests such as grip strength assessment. 3. Improvement in respiratory function: – Assessment of respiratory muscle strength and function using pulmonary function tests (e.g., forced vital capacity (FVC), peak cough flow) to monitor changes in respiratory function and breathing capacity. 4. Reduction in disease progression: – Evaluation of disease progression through longitudinal assessment of motor function, respiratory function, and overall clinical status over time. 5. Increase in survival rates and disease-free intervals: – Monitoring of overall survival rates and disease-free intervals following treatment with cellular therapy and stem cells. 6. Improvement in quality of life: – Utilization of validated quality of life measures specific to SMA, such as the Pediatric Quality of Life Inventory (PedsQL) or the SMA Quality of Life questionnaire (SMA-QoL), to assess changes in physical, emotional, and social well-being. Consult with Our Team of Experts Now!
|
1.20 Transverse Myelitis (TM) 1.20.2 Chronic Transverse Myelitis 1.20.3 Recurrent Transverse Myelitis 1.20.4 Post-infectious Transverse Myelitis 1.20.5 Idiopathic Transverse Myelitis 1.20.6 Neuromyelitis Optica Spectrum Disorder (NMOSD)-associated Transverse Myelitis 1.20.7 Multiple Sclerosis (MS)-related Transverse Myelitis |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Improvement in motor function: – Assessment using standardized motor scales such as the Medical Research Council (MRC) scale or the Modified Rankin Scale (mRS) to evaluate changes in muscle strength, coordination, and motor control. 2. Enhancement of sensory function: – Evaluation of sensory modalities (light touch, pinprick, proprioception) using standardized sensory scales to assess improvements in sensory perception, discrimination, and neuropathic pain. 3. Reduction in spasticity: – Measurement of spasticity using scales such as the Modified Ashworth Scale (MAS) to quantify changes in muscle tone and spasticity levels. 4. Improvement in bladder and bowel function: – Assessment of bladder and bowel continence, voiding patterns, and bowel movements to determine improvements in bladder and bowel function post-treatment. 5. Preservation or restoration of ambulation: – Evaluation of walking ability, gait parameters, and mobility using standardized tests such as the Timed Up and Go (TUG) test or the 10-Meter Walk Test (10MWT). 6. Reduction in neuropathic pain: – Assessment of neuropathic pain using pain scales (e.g., visual analog scale (VAS), Numerical Rating Scale (NRS)) to quantify changes in pain intensity and frequency. 7. Improvement in quality of life: – Utilization of validated quality of life measures specific to Transverse Myelitis, such as the Transverse Myelitis Quality of Life questionnaire (TM-QOL) or the Short Form Health Survey (SF-36), to assess changes in physical, emotional, and social well-being. Consult with Our Team of Experts Now!
|
1.21 Traumatic Brain Injury (TBI) 1.21.1 Mild Traumatic Brain Injury (mTBI) or Concussion 1.21.2 Moderate Traumatic Brain Injury 1.21.3 Severe Traumatic Brain Injury 1.21.4 Penetrating Traumatic Brain Injury 1.21.5 Closed Traumatic Brain Injury 1.21.6 Diffuse Axonal Injury (DAI) 1.21.7 Focal Brain Injury 1.21.8 Blast-Related Traumatic Brain Injury |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Improvement in neurological function: – Assessment using standardized neurological scales such as the Glasgow Coma Scale (GCS) or the Rancho Los Amigos Levels of Cognitive Functioning Scale to evaluate changes in consciousness, cognitive function, and neurological deficits. 2. Reduction in intracranial pressure (ICP) and cerebral edema: – Monitoring of ICP levels through invasive or non-invasive techniques to assess changes in cerebral perfusion and edema post-treatment. 3. Preservation or improvement in cognitive function: – Evaluation using neuropsychological tests to assess changes in memory, attention, executive function, language, and visuospatial abilities. 4. Restoration of motor function: – Assessment using standardized motor scales such as the Motor Assessment Scale (MAS) or the Functional Independence Measure (FIM) to evaluate changes in muscle strength, coordination, and functional abilities. 5. Improvement in functional independence: – Measurement of activities of daily living (ADLs) using scales such as the Functional Independence Measure (FIM) or the Barthel Index to assess changes in functional status and independence in daily tasks. 6. Reduction in post-traumatic seizures: – Monitoring of seizure activity through electroencephalography (EEG) or clinical assessment to evaluate changes in seizure frequency and severity post-treatment. 7. Increase in quality of life: – Utilization of validated quality of life measures specific to TBI, such as the Quality of Life after Brain Injury (QOLIBRI) or the Short Form Health Survey (SF-36), to assess changes in physical, emotional, and social well-being. Consult with Our Team of Experts Now!
|
1.22 Brain tumors 1.22.1 Glioblastoma multiforme (GBM) 1.22.2 Astrocytoma (Grade I-IV) 1.22.3 Oligodendroglioma 1.22.4 Ependymoma 1.22.5 Medulloblastoma 1.22.6 Meningioma 1.22.7 Pituitary adenoma 1.22.8 Craniopharyngioma 1.22.9 Pineal gland tumors 1.22.10 Schwannoma 1.22.11 Central nervous system lymphoma 1.22.12 Metastatic brain tumors |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Reduction in tumor size and volume: – Evaluation using neuroimaging techniques such as MRI or CT scans to measure changes in tumor size, volume, and enhancement patterns post-treatment. 2. Improvement in neurological function: – Assessment using standardized neurological scales such as the Karnofsky Performance Status (KPS) or the World Health Organization Performance Status (WHO PS) to evaluate changes in neurological deficits, cognitive function, and functional status. 3. Preservation or improvement in cognitive function: – Evaluation using neuropsychological tests to assess changes in memory, attention, executive function, language, and visuospatial abilities. 4. Reduction in tumor-related symptoms: – Assessment of tumor-related symptoms such as headaches, seizures, focal neurological deficits, and changes in mental status to determine improvements in symptom control and overall quality of life. 5. Increase in progression-free survival (PFS) and overall survival (OS) rates: – Monitoring of disease progression and survival rates over time to assess the efficacy of cellular therapy and stem cells in prolonging survival and delaying disease progression. 6. Reduction in treatment-related side effects: – Evaluation of treatment-related side effects such as neurotoxicity, immunosuppression, and hematological toxicity to determine the safety profile of cellular therapy and stem cells in patients with brain tumors. Consult with Our Team of Experts Now!
|
1.23 Rett syndrome 1.23.1 Typical Rett Syndrome (Classic Rett Syndrome) 1.23.2 Atypical Rett Syndrome 1.23.3 Zappella Variant Rett Syndrome 1.23.4 Hanefeld Variant Rett Syndrome 1.23.5 Congenital Variant Rett Syndrome |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Improvement in motor function: – Assessment using standardized motor scales such as the Rett Syndrome Motor-Behavioral Assessment Battery (RSM-BAB) or the Rett Syndrome Gross Motor Scale (RSGMS) to evaluate changes in gross motor function, coordination, and motor skills. 2. Enhancement of communication skills: – Evaluation of communication abilities using standardized communication scales such as the Communication and Symbolic Behavior Scales (CSBS) or the Vineland Adaptive Behavior Scales (VABS) to assess changes in expressive and receptive language skills. 3. Reduction in stereotypic behaviors: – Measurement of stereotypic behaviors and repetitive movements using standardized behavioral scales or observational tools to determine changes in behavior patterns and symptom severity. 4. Improvement in cognitive function: – Assessment using standardized cognitive scales such as the Bayley Scales of Infant and Toddler Development (Bayley-III) or the Mullen Scales of Early Learning to evaluate changes in cognitive abilities, attention, and problem-solving skills. 5. Enhancement of social interaction and engagement: – Evaluation of social skills and interaction using standardized socialization scales such as the Social Responsiveness Scale (SRS) or the Social Communication Questionnaire (SCQ) to assess changes in social behavior and engagement with others. 6. Reduction in seizures: – Monitoring of seizure frequency, duration, and severity using seizure diaries or electroencephalography (EEG) to determine changes in seizure control and management post-treatment. 7. Improvement in quality of life: – Utilization of validated quality of life measures specific to Rett syndrome, such as the Rett Syndrome Quality of Life questionnaire (RS-QOL) or the Pediatric Quality of Life Inventory (PedsQL), to assess changes in physical, emotional, and social well-being. Consult with Our Team of Experts Now!
|
1.24 Schizophrenia |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Symptom Severity – Positive Symptoms: These include hallucinations, delusions, and thought disorders. Tools like the Positive and Negative Syndrome Scale (PANSS) or the Brief Psychiatric Rating Scale (BPRS) are often used. – Negative Symptoms: These include affective flattening, alogia, and avolition. The PANSS and the Scale for the Assessment of Negative Symptoms (SANS) are commonly used. 2. Functional Outcomes – Social Functioning: Assessed through tools like the Global Assessment of Functioning (GAF) scale or the Social Functioning Scale (SFS), which evaluate the ability to engage in social interactions and maintain relationships. – Occupational Functioning: Evaluated based on the patient’s ability to maintain employment or engage in meaningful activities. – Daily Living Skills: Assessed by evaluating the patient’s ability to perform everyday tasks, often using tools like the Independent Living Skills Survey (ILSS). 3. Quality of Life – Tools like the Quality of Life Scale (QLS) or the WHOQOL-BREF are used to measure the patient’s overall well-being and satisfaction with life across various domains, such as physical health, psychological state, social relationships, and environment. 4. Cognitive Functioning – Neurocognitive Assessments: These assess various cognitive domains including memory, attention, executive function, and processing speed. Tools such as the MATRICS Consensus Cognitive Battery (MCCB) are often used. 5. Relapse Rates and Hospitalization – Monitoring the frequency and duration of relapses, as well as the need for hospitalization, is crucial for understanding the long-term stability and effectiveness of treatment. 6. Adverse Effects of Treatment – Medication Side Effects: Evaluated using scales like the Simpson-Angus Scale (SAS) for extrapyramidal symptoms, the Abnormal Involuntary Movement Scale (AIMS) for tardive dyskinesia, and the Barnes Akathisia Rating Scale (BARS) for akathisia. – Metabolic Side Effects: Monitoring weight, glucose levels, and lipid profiles, especially in patients on antipsychotic medications, which can have significant metabolic effects. 7. Adherence to Treatment – Evaluated through patient self-reports, pill counts, or electronic monitoring to ensure that patients are following their prescribed treatment regimens. Consult with Our Team of Experts Now!
|
1.25 Bipolar disorder |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Mood Episode Severity and Frequency – Manic Episodes: Assessed using tools like the Young Mania Rating Scale (YMRS) or the Mania Rating Scale (MRS). – Depressive Episodes: Evaluated with scales such as the Hamilton Depression Rating Scale (HDRS) or the Montgomery-Åsberg Depression Rating Scale (MADRS). – Mixed Episodes: Assessed with tools that can capture both manic and depressive symptoms, like the Bipolar Inventory of Symptoms Scale (BISS). 2. Functional Outcomes – Social Functioning: Assessed through tools like the Social Adjustment Scale (SAS) or the Social Functioning Scale (SFS), which evaluate the patient’s ability to engage in social interactions and maintain relationships. – Occupational Functioning: Evaluated based on the patient’s ability to maintain employment or engage in meaningful activities. – Daily Living Skills: Assessed by evaluating the patient’s ability to perform everyday tasks, often using tools like the Independent Living Skills Survey (ILSS). 3. Quality of Life – Tools like the Quality of Life in Bipolar Disorder (QoL.BD) scale or the WHOQOL-BREF are used to measure the patient’s overall well-being and satisfaction with life across various domains, such as physical health, psychological state, social relationships, and environment. 4. Cognitive Functioning – Neurocognitive Assessments: These assess various cognitive domains including memory, attention, executive function, and processing speed. Tools such as the MATRICS Consensus Cognitive Battery (MCCB) or specific tests for bipolar disorder are often used. 5. Relapse Rates and Hospitalization – Monitoring the frequency and duration of manic, depressive, and mixed episodes, as well as the need for hospitalization, is crucial for understanding long-term stability and effectiveness of treatment. 6. Adverse Effects of Treatment – Medication Side Effects: Evaluated using scales like the Systematic Assessment for Treatment Emergent Events (SAFTEE) or the Side Effect Rating Scale (SERS). – Metabolic Side Effects: Monitoring weight, glucose levels, and lipid profiles, especially in patients on mood stabilizers or antipsychotic medications, which can have significant metabolic effects. 7. Adherence to Treatment – Evaluated through patient self-reports, pill counts, or electronic monitoring to ensure that patients are following their prescribed treatment regimens. 8. Suicide Risk – Assessed using tools like the Columbia-Suicide Severity Rating Scale (C-SSRS) or the Beck Scale for Suicide Ideation (BSS) to evaluate the risk of self-harm or suicidal behavior. Consult with Our Team of Experts Now!
|
1.26 Major depressive disorder |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Symptom Severity – Depressive Symptoms: Assessed using tools such as the Hamilton Depression Rating Scale (HAM-D), the Montgomery-Åsberg Depression Rating Scale (MADRS), or the Beck Depression Inventory (BDI). – Anxiety Symptoms: Often evaluated as co-occurring symptoms using tools like the Hamilton Anxiety Rating Scale (HAM-A). 2. Functional Outcomes – Social Functioning: Assessed through instruments like the Social Adjustment Scale (SAS) or the Social Functioning Scale (SFS), which evaluate the patient’s ability to engage in social interactions and maintain relationships. – Occupational Functioning: Evaluated based on the patient’s ability to maintain employment or engage in meaningful activities. – Daily Living Skills: Assessed by evaluating the patient’s ability to perform everyday tasks, often using tools like the Sheehan Disability Scale (SDS). 3. Quality of Life – Tools such as the Quality of Life Enjoyment and Satisfaction Questionnaire (Q-LES-Q) or the WHOQOL-BREF are used to measure the patient’s overall well-being and satisfaction with life across various domains, including physical health, psychological state, social relationships, and environment. 4. Cognitive Functioning – Neurocognitive Assessments: These assess various cognitive domains including memory, attention, executive function, and processing speed. Cognitive impairment is often evaluated using tools like the Cognitive Functioning Subscale of the HAM-D. 5. Relapse Rates and Remission – Relapse Rates: Monitoring the frequency and duration of depressive episodes to understand long-term stability and effectiveness of treatment. – Remission Rates: Evaluating the extent to which patients achieve and maintain symptom-free periods. Remission is often defined as a specific score threshold on depression rating scales (e.g., HAM-D ≤ 7). 6. Adverse Effects of Treatment – Medication Side Effects: Evaluated using scales like the Systematic Assessment for Treatment Emergent Events (SAFTEE) or the Antidepressant Side-Effect Checklist (ASEC). – Metabolic Side Effects: Monitoring weight, glucose levels, and lipid profiles, particularly in patients on antidepressants that can have metabolic effects. 7. Adherence to Treatment – Evaluated through patient self-reports, pill counts, or electronic monitoring to ensure that patients are following their prescribed treatment regimens. 8. Suicide Risk – Assessed using tools such as the Columbia-Suicide Severity Rating Scale (C-SSRS) or the Beck Scale for Suicide Ideation (BSS) to evaluate the risk of self-harm or suicidal behavior. Consult with Our Team of Experts Now!
|
1.27 Friedreich’s ataxia |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Neurological Symptoms – Ataxia Severity: Assessed using scales such as the Friedreich Ataxia Rating Scale (FARS), which evaluates coordination, speech, and gait disturbances. – Dysarthria: Evaluated using specific speech assessment tools to measure speech difficulties, a common symptom in FRDA. – Peripheral Neuropathy: Assessed using clinical examinations and tools like nerve conduction studies. 2. Functional Outcomes – Gait and Mobility: Assessed using tools such as the Timed 25-Foot Walk (T25FW) or the 6-Minute Walk Test (6MWT) to measure walking ability and endurance. – Upper Limb Function: Evaluated using tests like the 9-Hole Peg Test (9HPT) to assess fine motor skills and hand coordination. – Activities of Daily Living: Assessed using scales like the Functional Independence Measure (FIM) or the Barthel Index to evaluate the patient’s ability to perform everyday tasks. 3. Cardiomyopathy – Cardiac Function: Monitored through echocardiography, electrocardiograms (ECG), and cardiac MRI to evaluate heart size, function, and structure, as cardiomyopathy is a common complication of FRDA. 4. Diabetes and Glucose Metabolism – Blood Glucose Levels: Regular monitoring of fasting glucose and HbA1c levels to assess for diabetes, which is a common comorbidity in FRDA patients. 5. Skeletal Deformities – Scoliosis: Monitored through clinical examinations and radiographs to assess the progression of spinal curvature. – Foot Deformities: Assessed through physical examination and imaging studies. 6. Quality of Life – Tools such as the SF-36 Health Survey or the Friedreich Ataxia Impact Scale (FAIS) are used to measure the patient’s overall well-being and satisfaction with life across various domains, including physical health, psychological state, social relationships, and environment. 7. Genetic and Biomarker Assessments – Genetic Testing: Used to confirm the diagnosis of FRDA by identifying mutations in the FXN gene. – Biomarkers: Research is ongoing to identify biomarkers for disease progression and treatment response. 8. Adverse Effects of Treatment – Medication Side Effects: Evaluated through patient self-reports and clinical monitoring to ensure safety and tolerability of treatments. – Physical Therapy and Rehabilitation: Assessment of response to therapeutic interventions aimed at improving or maintaining function. 9. Cognitive Functioning – Neurocognitive Assessments: These assess various cognitive domains including memory, attention, executive function, and processing speed, although cognitive impairment is less common in FRDA compared to other symptoms. Consult with Our Team of Experts Now!
|
1.28 Leukodystrophies |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Neurological Symptoms – Motor Function: Assessed using scales like the Gross Motor Function Classification System (GMFCS) and the Hammersmith Functional Motor Scale (HFMS) to evaluate muscle tone, strength, coordination, and gait. – Spasticity: Evaluated using tools such as the Modified Ashworth Scale (MAS) to measure muscle stiffness and spasticity. – Seizure Activity: Monitored through clinical evaluations and electroencephalograms (EEG). 2. Cognitive Function – Cognitive Assessments: Tools like the Wechsler Intelligence Scale for Children (WISC) or the Bayley Scales of Infant Development are used to assess cognitive function in children, while tools like the Mini-Mental State Examination (MMSE) may be used for adults. 3. Functional Outcomes – Daily Living Skills: Assessed using scales like the Functional Independence Measure (FIM) or the Pediatric Evaluation of Disability Inventory (PEDI) to evaluate the patient’s ability to perform everyday tasks. – Speech and Language: Evaluated through speech and language assessments to measure communication abilities and speech clarity. 4. Disease Progression – MRI and Imaging Studies: Regular MRI scans are used to monitor changes in white matter and assess disease progression. – Biomarkers: Research is ongoing to identify biomarkers that can help track disease progression and response to treatment. 5. Quality of Life – Quality of Life Assessments: Tools such as the Pediatric Quality of Life Inventory (PedsQL) or the SF-36 Health Survey are used to measure the patient’s overall well-being and satisfaction with life across various domains, including physical health, psychological state, social relationships, and environment. 6. Growth and Development – Growth Charts: Regular monitoring of height, weight, and head circumference to assess growth and development in children. – Developmental Milestones: Assessed using tools like the Denver Developmental Screening Test (DDST) to evaluate the achievement of age-appropriate developmental milestones. 7. Vision and Hearing – Vision Assessments: Regular eye exams to monitor visual acuity and detect any vision problems. – Hearing Assessments: Audiometric testing to evaluate hearing function and identify hearing loss. 8. Pain and Comfort – Pain Assessment: Tools like the Wong-Baker FACES Pain Rating Scale or the Numeric Pain Rating Scale (NPRS) are used to assess pain levels and discomfort. – Comfort Measures: Evaluated through patient and caregiver reports to ensure optimal comfort and quality of life. 9. Adverse Effects of Treatment – Medication Side Effects: Monitored through patient self-reports and clinical evaluations to ensure the safety and tolerability of treatments. – Therapeutic Interventions: Assessment of response to physical, occupational, and speech therapy aimed at improving or maintaining function. 10. Genetic and Biomarker Assessments – Genetic Testing: Used to confirm the diagnosis of specific leukodystrophies by identifying mutations in relevant genes. – Biomarkers: Ongoing research to identify biomarkers for disease progression and treatment response. Consult with Our Team of Experts Now!
|
1.29 Ischemic brain injury |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Neurological Function – Stroke Severity: Assessed using scales such as the National Institutes of Health Stroke Scale (NIHSS) to evaluate the extent of neurological impairment. – Motor Function: Evaluated using tools like the Fugl-Meyer Assessment (FMA) to assess motor recovery in the affected limbs. – Sensory Function: Assessed through clinical examinations to evaluate sensory deficits. 2. Functional Outcomes – Activities of Daily Living (ADL): Assessed using tools like the Barthel Index or the Functional Independence Measure (FIM) to evaluate the patient’s ability to perform daily tasks independently. – Mobility and Gait: Evaluated using the Timed Up and Go (TUG) test or the 6-Minute Walk Test (6MWT) to assess walking ability and endurance. 3. Cognitive Function – Cognitive Assessments: Tools such as the Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment (MoCA), or the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) are used to evaluate various cognitive domains including memory, attention, executive function, and language. 4. Quality of Life – Quality of Life Assessments: Instruments like the Stroke Impact Scale (SIS) or the SF-36 Health Survey are used to measure the patient’s overall well-being and satisfaction with life across various domains, including physical health, psychological state, social relationships, and environment. 5. Psychological and Emotional Health – Depression and Anxiety: Assessed using tools like the Patient Health Questionnaire-9 (PHQ-9) for depression and the Generalized Anxiety Disorder 7 (GAD-7) scale for anxiety. – Emotional and Behavioral Changes: Evaluated through clinical interviews and behavioral assessments. 6. Speech and Language – Aphasia Severity: Assessed using tools like the Boston Diagnostic Aphasia Examination (BDAE) or the Western Aphasia Battery (WAB) to evaluate speech and language impairments. – Dysarthria: Evaluated using speech assessments to measure speech clarity and articulation difficulties. 7. Swallowing Function – Dysphagia Assessment: Assessed using clinical bedside evaluations and instrumental assessments like videofluoroscopic swallow studies (VFSS) or fiberoptic endoscopic evaluation of swallowing (FEES). 8. Stroke Recurrence and Secondary Prevention – Recurrence Rates: Monitoring the incidence of recurrent strokes to understand long-term stability and effectiveness of secondary prevention measures. – Risk Factor Management: Evaluated through clinical monitoring of blood pressure, lipid levels, glucose levels, and lifestyle modifications. 9. Adverse Effects of Treatment – Medication Side Effects: Monitored through patient self-reports and clinical evaluations to ensure the safety and tolerability of treatments such as anticoagulants, antiplatelets, and antihypertensives. – Rehabilitation Interventions: Assessment of response to physical, occupational, and speech therapy aimed at improving or maintaining function. 10. Imaging and Biomarkers – Neuroimaging: Regular MRI or CT scans to monitor brain structure and the extent of ischemic damage. – Biomarkers: Research is ongoing to identify biomarkers that can help track disease progression and response to treatment. Consult with Our Team of Experts Now!
|
1.30 Retinal diseases (e.g., macular degeneration, retinitis pigmentosa) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Visual Function – Visual Acuity: Assessed using the Snellen chart or the Early Treatment Diabetic Retinopathy Study (ETDRS) chart to measure central vision sharpness. – Contrast Sensitivity: Evaluated using tests like the Pelli-Robson chart to assess the ability to distinguish between different shades of grey. – Visual Field Testing: Conducted using perimetry tests, such as the Humphrey Field Analyzer, to evaluate peripheral vision and detect scotomas (blind spots). – Color Vision: Assessed using tests like the Ishihara plates or the Farnsworth-Munsell 100 hue test to evaluate color discrimination. 2. Disease Progression – Retinal Imaging: Regularly performed using optical coherence tomography (OCT) to monitor structural changes in the retina, such as retinal thickness and the presence of fluid or atrophy. – Fundus Photography: Used to document changes in the retina over time, including drusen in macular degeneration or pigmentary changes in retinitis pigmentosa. – Fluorescein Angiography (FA): Performed to visualize the blood vessels in the retina and detect abnormalities such as leakage or neovascularization. 3. Functional Outcomes – Reading Ability: Assessed using reading charts or specific tests designed to evaluate reading speed and comprehension, important for daily functioning. – Activities of Daily Living (ADL): Evaluated using scales like the National Eye Institute Visual Function Questionnaire (NEI VFQ-25) to assess the impact of vision loss on daily activities. 4. Quality of Life – Health-Related Quality of Life (HRQoL): Measured using instruments like the NEI VFQ-25 or the Impact of Vision Impairment (IVI) questionnaire to evaluate the patient’s overall well-being and satisfaction with life. – Emotional and Psychological Health: Assessed using tools like the Hospital Anxiety and Depression Scale (HADS) or the Geriatric Depression Scale (GDS) to evaluate the impact of vision loss on mental health. 5. Mobility and Independence – Orientation and Mobility Skills: Evaluated through assessments of the patient’s ability to navigate different environments safely and independently. – Use of Assistive Devices: Monitored to understand the patient’s use and effectiveness of low-vision aids, such as magnifiers, screen readers, or mobility canes. 6. Treatment Side Effects and Adherence – Medication Side Effects: Monitored through patient self-reports and clinical evaluations to ensure the safety and tolerability of treatments, such as anti-VEGF injections for macular degeneration. – Adherence: Evaluated through patient self-reports, medication logs, or electronic monitoring to ensure compliance with prescribed treatment regimens. 7. Genetic and Biomarker Assessments – Genetic Testing: Used to confirm the diagnosis of specific retinal diseases by identifying mutations in relevant genes, particularly for inherited conditions like retinitis pigmentosa. – Biomarkers: Research is ongoing to identify biomarkers in blood or ocular fluids that can help track disease progression and response to treatment. 8. Pain and Discomfort – Ocular Pain Assessment: Tools like the Numeric Pain Rating Scale (NPRS) or specific questionnaires for ocular discomfort are used to assess pain levels and discomfort associated with retinal diseases or treatments. 9. Social and Occupational Impact – Social Functioning: Assessed using tools like the Lubben Social Network Scale (LSNS) to evaluate social connections and support. – Occupational Impact: Evaluated through assessments of the patient’s ability to maintain employment or engage in meaningful activities despite vision loss. 10. Coping Strategies and Rehabilitation – Adaptive Coping Strategies: Evaluated through interviews and questionnaires to understand how patients cope with vision loss and what strategies they use to adapt. – Rehabilitation Outcomes: Assessed through the effectiveness of vision rehabilitation programs, including training in the use of assistive devices and environmental modifications. Consult with Our Team of Experts Now!
|
1.31 Leber’s hereditary optic neuropathy (LHON) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Visual Function – Visual Acuity: Assessed using standard eye charts such as the Snellen chart or the Early Treatment Diabetic Retinopathy Study (ETDRS) chart to measure central vision sharpness. – Visual Field Testing: Conducted using automated perimetry tests, such as the Humphrey Field Analyzer, to evaluate peripheral vision and detect central scotomas (blind spots). – Color Vision: Evaluated using tests like the Ishihara plates or the Farnsworth-Munsell 100 hue test to assess color discrimination. – Contrast Sensitivity: Assessed using charts like the Pelli-Robson contrast sensitivity chart to measure the ability to distinguish between different shades of grey. 2. Disease Progression – Optical Coherence Tomography (OCT): Regularly performed to monitor structural changes in the optic nerve and retinal nerve fiber layer (RNFL) thickness. – Fundus Photography: Used to document changes in the optic disc and retinal appearance over time. – Visual Evoked Potentials (VEP): Conducted to measure the electrical activity in the brain in response to visual stimuli, providing information on the functional status of the visual pathways. 3. Functional Outcomes – Reading Ability: Assessed using reading charts or specific tests designed to evaluate reading speed and comprehension, which are crucial for daily functioning. – Activities of Daily Living (ADL): Evaluated using scales like the National Eye Institute Visual Function Questionnaire (NEI VFQ-25) to assess the impact of vision loss on daily activities. 4. Quality of Life – Health-Related Quality of Life (HRQoL): Measured using instruments like the NEI VFQ-25 or the Impact of Vision Impairment (IVI) questionnaire to evaluate the patient’s overall well-being and satisfaction with life. – Emotional and Psychological Health: Assessed using tools like the Hospital Anxiety and Depression Scale (HADS) or the Geriatric Depression Scale (GDS) to evaluate the impact of vision loss on mental health. 5. Genetic and Biomarker Assessments – Genetic Testing: Used to confirm the diagnosis of LHON by identifying mutations in mitochondrial DNA, particularly the common mutations (e.g., m.11778G>A, m.14484T>C, and m.3460G>A). – Biomarkers: Research is ongoing to identify biomarkers in blood or ocular fluids that can help track disease progression and response to treatment. 6. Treatment Side Effects and Adherence – Medication Side Effects: Monitored through patient self-reports and clinical evaluations to ensure the safety and tolerability of treatments, such as idebenone or other experimental therapies. – Adherence: Evaluated through patient self-reports, medication logs, or electronic monitoring to ensure compliance with prescribed treatment regimens. 7. Mobility and Independence – Orientation and Mobility Skills: Evaluated through assessments of the patient’s ability to navigate different environments safely and independently. – Use of Assistive Devices: Monitored to understand the patient’s use and effectiveness of low-vision aids, such as magnifiers, screen readers, or mobility canes. 8. Social and Occupational Impact – Social Functioning: Assessed using tools like the Lubben Social Network Scale (LSNS) to evaluate social connections and support. – Occupational Impact: Evaluated through assessments of the patient’s ability to maintain employment or engage in meaningful activities despite vision loss. 9. Pain and Discomfort – Ocular Pain Assessment: Tools like the Numeric Pain Rating Scale (NPRS) or specific questionnaires for ocular discomfort are used to assess pain levels and discomfort associated with LHON or treatments. 10. Coping Strategies and Rehabilitation – Adaptive Coping Strategies: Evaluated through interviews and questionnaires to understand how patients cope with vision loss and what strategies they use to adapt. – Rehabilitation Outcomes: Assessed through the effectiveness of vision rehabilitation programs, including training in the use of assistive devices and environmental modifications. Consult with Our Team of Experts Now!
|
1.32 Ataxia telangiectasia (AT) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Neurological Function – Ataxia Severity: Assessed using scales such as the International Cooperative Ataxia Rating Scale (ICARS) or the Scale for the Assessment and Rating of Ataxia (SARA) to evaluate coordination, gait, and balance. – Dysarthria: Evaluated using specific speech assessment tools to measure speech difficulties. – Oculomotor Abnormalities: Assessed through clinical evaluations of eye movements to detect problems such as nystagmus or difficulty with saccades. – Peripheral Neuropathy: Assessed using clinical examinations and tools like nerve conduction studies. 2. Functional Outcomes – Activities of Daily Living (ADL): Assessed using scales like the Barthel Index or the Functional Independence Measure (FIM) to evaluate the patient’s ability to perform daily tasks independently. – Mobility and Gait: Evaluated using the Timed Up and Go (TUG) test or the 6-Minute Walk Test (6MWT) to assess walking ability and endurance. – Upper Limb Function: Assessed using tests like the 9-Hole Peg Test (9HPT) to evaluate fine motor skills and hand coordination. 3. Immunological Status – Immune Function Tests: Regular blood tests to assess immunoglobulin levels (IgA, IgG, IgM) and lymphocyte subpopulations to monitor immune system function. – Infection History: Documenting the frequency, severity, and types of infections to evaluate immune competence. 4. Cancer Risk – Screening for Malignancies: Regular screenings and imaging studies to detect early signs of cancers, particularly lymphomas and leukemias, which are more common in A-T patients. 5. Respiratory Function – Pulmonary Function Tests (PFTs): Regular spirometry to assess lung function and detect respiratory complications. – Respiratory Infections: Monitoring the frequency and severity of respiratory infections to manage and prevent complications. 6. Growth and Development – Growth Charts: Regular monitoring of height, weight, and head circumference to assess growth and development in children. – Developmental Milestones: Assessed using tools like the Denver Developmental Screening Test (DDST) to evaluate the achievement of age-appropriate developmental milestones. 7. Quality of Life – Health-Related Quality of Life (HRQoL): Measured using instruments like the Pediatric Quality of Life Inventory (PedsQL) or the SF-36 Health Survey to evaluate the patient’s overall well-being and satisfaction with life. – Emotional and Psychological Health: Assessed using tools like the Geriatric Depression Scale (GDS) or the Patient Health Questionnaire-9 (PHQ-9) to evaluate the impact of the disease on mental health. 8. Genetic and Biomarker Assessments – Genetic Testing: Used to confirm the diagnosis of A-T by identifying mutations in the ATM gene. – Biomarkers: Research is ongoing to identify biomarkers that can help track disease progression and response to treatment. 9. Treatment Side Effects and Adherence – Medication Side Effects: Monitored through patient self-reports and clinical evaluations to ensure the safety and tolerability of treatments, such as immunoglobulin replacement therapy or steroids. – Adherence: Evaluated through patient self-reports, medication logs, or electronic monitoring to ensure compliance with prescribed treatment regimens. 10. Skeletal and Skin Abnormalities – Telangiectasias: Clinical evaluation of characteristic telangiectasias (small dilated blood vessels) typically seen on the eyes, ears, and sun-exposed areas of the skin. – Scoliosis and Other Skeletal Issues: Monitored through clinical examinations and radiographs to assess the progression of spinal curvature and other skeletal deformities. 11. Pain and Comfort – Pain Assessment: Tools like the Wong-Baker FACES Pain Rating Scale or the Numeric Pain Rating Scale (NPRS) are used to assess pain levels and discomfort. – Comfort Measures: Evaluated through patient and caregiver reports to ensure optimal comfort and quality of life. 12. Social and Occupational Impact – Social Functioning: Assessed using tools like the Lubben Social Network Scale (LSNS) to evaluate social connections and support. – Occupational and Educational Impact: Evaluated through assessments of the patient’s ability to engage in meaningful activities, education, and employment despite the disease. Consult with Our Team of Experts Now!
|
1.33 Neuronal ceroid lipofuscinosis (NCL) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Neurological Function – Cognitive Function: Assessed using tools such as the Wechsler Intelligence Scale for Children (WISC) or the Bayley Scales of Infant and Toddler Development to evaluate intellectual abilities. – Motor Function: Evaluated through clinical examinations and scales like the Gross Motor Function Classification System (GMFCS) to assess muscle tone, strength, coordination, and gait. – Seizure Activity: Monitored through clinical evaluations and electroencephalograms (EEG). 2. Visual Function – Visual Acuity: Assessed using eye charts like the Snellen chart or specific tests for children to measure central vision sharpness. – Visual Field Testing: Conducted using perimetry tests to assess peripheral vision. – Retinal Imaging: Utilized to monitor changes in the retina over time, such as pigmentary changes or retinal thinning. 3. Functional Outcomes – Activities of Daily Living (ADL): Assessed using scales like the Pediatric Evaluation of Disability Inventory (PEDI) to evaluate the patient’s ability to perform daily tasks independently. – Mobility and Gait: Evaluated using tests like the Timed Up and Go (TUG) test or the 6-Minute Walk Test (6MWT) to assess walking ability and endurance. 4. Quality of Life – Health-Related Quality of Life (HRQoL): Measured using instruments such as the Pediatric Quality of Life Inventory (PedsQL) or disease-specific quality of life measures to assess the impact of NCL on the patient’s overall well-being and satisfaction with life. – Emotional and Psychological Health: Assessed using tools like the Child Behavior Checklist (CBCL) or the Strengths and Difficulties Questionnaire (SDQ) to evaluate behavioral and emotional functioning. 5. Genetic and Biomarker Assessments – Genetic Testing: Used to confirm the diagnosis of NCL and identify specific mutations in genes associated with different subtypes of the disease (e.g., CLN1, CLN2, CLN3, etc.). – Biomarkers: Research is ongoing to identify biomarkers in blood or cerebrospinal fluid (CSF) that can help track disease progression and response to treatment. 6. Respiratory Function – Pulmonary Function Tests (PFTs): Regular spirometry to assess lung function and detect respiratory complications, which may occur as the disease progresses. 7. Swallowing and Nutritional Status – Dysphagia Assessment: Evaluated through clinical examinations and assessments to monitor swallowing difficulties and ensure adequate nutrition and hydration. 8. Seizure Management – Seizure Control: Monitoring the frequency, severity, and response to antiepileptic medications to manage seizures effectively. 9. Treatment Side Effects and Adherence – Medication Side Effects: Monitored through patient self-reports and clinical evaluations to ensure the safety and tolerability of treatments, such as antiepileptic drugs or supportive care measures. – Adherence: Evaluated through patient and caregiver reports to ensure compliance with prescribed treatment regimens. 10. Social and Family Impact – Social Functioning: Assessed using tools like the Family Impact Scale (FIS) or the Pediatric Quality of Life Family Impact Module to evaluate the impact of NCL on family dynamics and social interactions. Consult with Our Team of Experts Now!
|
1.34 Canavan disease |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Neurological Function – Motor Function: Evaluated through clinical examinations and scales like the Gross Motor Function Classification System (GMFCS) to assess muscle tone, strength, coordination, and motor skills. – Cognitive Function: Assessed using developmental scales appropriate for the child’s age, such as the Bayley Scales of Infant and Toddler Development or the Vineland Adaptive Behavior Scales, to evaluate intellectual abilities and developmental milestones. – Seizure Activity: Monitored through clinical evaluations and electroencephalograms (EEG) to assess seizure frequency and severity. 2. Communication and Language – Language Development: Assessed through standardized tests and observations to evaluate speech and language skills, which may be delayed or impaired in Canavan disease. 3. Visual and Hearing Function – Visual Acuity: Assessed using eye charts or vision tests to evaluate central vision sharpness. – Hearing: Evaluated through auditory tests to monitor hearing abilities and detect any impairment. 4. Functional Outcomes – Activities of Daily Living (ADL): Assessed using scales like the Pediatric Evaluation of Disability Inventory (PEDI) to evaluate the child’s ability to perform daily tasks independently. – Mobility and Gait: Evaluated using tests like the Timed Up and Go (TUG) test or the 6-Minute Walk Test (6MWT) to assess mobility and endurance. 5. Quality of Life – Health-Related Quality of Life (HRQoL): Measured using instruments such as the Pediatric Quality of Life Inventory (PedsQL) or disease-specific quality of life measures to assess the impact of Canavan disease on the child’s overall well-being and the family’s quality of life. – Emotional and Psychological Health: Assessed through caregiver reports and tools like the Pediatric Symptom Checklist (PSC) to evaluate behavioral and emotional functioning. 6. Genetic and Biomarker Assessments – Genetic Testing: Used to confirm the diagnosis of Canavan disease by identifying mutations in the ASPA gene. – Biomarkers: Research is ongoing to identify biomarkers in blood or cerebrospinal fluid (CSF) that can help track disease progression and response to treatment. 7. Nutritional Status and Swallowing Function – Dysphagia Assessment: Evaluated through clinical examinations and assessments to monitor swallowing difficulties and ensure adequate nutrition and hydration. 8. Seizure Management – Seizure Control: Monitoring the frequency, severity, and response to antiepileptic medications to manage seizures effectively. 9. Treatment Side Effects and Adherence – Medication Side Effects: Monitored through patient self-reports and clinical evaluations to ensure the safety and tolerability of treatments, such as antiepileptic drugs or supportive care measures. – Adherence: Evaluated through caregiver reports and medication logs to ensure compliance with prescribed treatment regimens. 10. Social and Family Impact – Social Functioning: Assessed using tools like the Family Impact Scale (FIS) or the Pediatric Quality of Life Family Impact Module to evaluate the impact of Canavan disease on family dynamics and social interactions. Consult with Our Team of Experts Now!
|
1.35 Sanfilippo syndrome |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Neurological Function – Cognitive Function: Assessed using developmental scales appropriate for the child’s age, such as the Bayley Scales of Infant and Toddler Development or the Vineland Adaptive Behavior Scales, to evaluate intellectual abilities and developmental milestones. – Motor Function: Evaluated through clinical examinations and scales like the Gross Motor Function Classification System (GMFCS) to assess muscle tone, strength, coordination, and motor skills. – Seizure Activity: Monitored through clinical evaluations and electroencephalograms (EEG) to assess seizure frequency and severity. 2. Communication and Language – Language Development: Assessed through standardized tests and observations to evaluate speech and language skills, which may be delayed or impaired in Sanfilippo syndrome. 3. Behavioral Changes – Behavioral Assessment: Evaluated through caregiver reports and standardized tools like the Child Behavior Checklist (CBCL) or the Aberrant Behavior Checklist (ABC) to assess behavioral changes, including hyperactivity, aggression, and sleep disturbances. 4. Functional Outcomes – Activities of Daily Living (ADL): Assessed using scales like the Pediatric Evaluation of Disability Inventory (PEDI) to evaluate the child’s ability to perform daily tasks independently. – Mobility and Gait: Evaluated using tests like the Timed Up and Go (TUG) test or the 6-Minute Walk Test (6MWT) to assess mobility and endurance. 5. Quality of Life – Health-Related Quality of Life (HRQoL): Measured using instruments such as the Pediatric Quality of Life Inventory (PedsQL) or disease-specific quality of life measures to assess the impact of Sanfilippo syndrome on the child’s overall well-being and the family’s quality of life. – Emotional and Psychological Health: Assessed through caregiver reports and tools like the Pediatric Symptom Checklist (PSC) to evaluate behavioral and emotional functioning. 6. Genetic and Biomarker Assessments – Genetic Testing: Used to confirm the diagnosis of Sanfilippo syndrome by identifying mutations in genes associated with MPS III (e.g., SGSH, NAGLU, HGSNAT, GNS). – Biomarkers: Research is ongoing to identify biomarkers in blood or cerebrospinal fluid (CSF) that can help track disease progression and response to treatment. 7. Nutritional Status and Swallowing Function – Dysphagia Assessment: Evaluated through clinical examinations and assessments to monitor swallowing difficulties and ensure adequate nutrition and hydration. 8. Seizure Management – Seizure Control: Monitoring the frequency, severity, and response to antiepileptic medications to manage seizures effectively. 9. Treatment Side Effects and Adherence – Medication Side Effects: Monitored through caregiver reports and clinical evaluations to ensure the safety and tolerability of treatments, such as enzyme replacement therapy or supportive care measures. – Adherence: Evaluated through caregiver reports and medication logs to ensure compliance with prescribed treatment regimens. 10. Social and Family Impact – Social Functioning: Assessed using tools like the Family Impact Scale (FIS) or the Pediatric Quality of Life Family Impact Module to evaluate the impact of Sanfilippo syndrome on family dynamics and social interactions. Consult with Our Team of Experts Now!
|
1.36 Metachromatic leukodystrophy (MLD) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Neurological Function – Motor Function: Evaluated through clinical examinations and scales such as the Gross Motor Function Classification System (GMFCS) to assess muscle tone, strength, coordination, and motor skills. – Cognitive Function: Assessed using developmental scales appropriate for the patient’s age, such as the Bayley Scales of Infant and Toddler Development or specific cognitive assessment tools to evaluate intellectual abilities and developmental milestones. – Speech and Language: Evaluated through standardized tests and observations to assess communication skills, which may become impaired as the disease progresses. 2. Functional Abilities – Activities of Daily Living (ADL): Assessed using scales like the Pediatric Evaluation of Disability Inventory (PEDI) or the Functional Independence Measure (FIM) to evaluate the patient’s ability to perform daily tasks independently. – Mobility and Gait: Evaluated using tests like the Timed Up and Go (TUG) test or the 6-Minute Walk Test (6MWT) to assess mobility, balance, and endurance. 3. Disease Progression – Neuroimaging: Utilized to monitor changes in the brain, particularly in the white matter, through techniques such as magnetic resonance imaging (MRI) to assess disease progression and structural abnormalities. – Electrophysiological Studies: Conducted to evaluate nerve conduction velocities and detect abnormalities in nerve function. 4. Quality of Life – Health-Related Quality of Life (HRQoL): Measured using instruments such as the Pediatric Quality of Life Inventory (PedsQL) or disease-specific quality of life measures to assess the impact of MLD on the patient’s overall well-being and the family’s quality of life. – Emotional and Psychological Health: Assessed through caregiver reports and tools like the Pediatric Symptom Checklist (PSC) or specific behavioral assessment scales to evaluate emotional functioning and behavioral changes. 5. Genetic and Biomarker Assessments – Genetic Testing: Used to confirm the diagnosis of MLD by identifying mutations in the ARSA gene (for the most common form) or other genes associated with variant forms. – Biomarkers: Research is ongoing to identify biomarkers in blood or cerebrospinal fluid (CSF) that can help track disease progression and response to treatment. 6. Respiratory Function – Pulmonary Function Tests (PFTs): Regular spirometry to assess lung function and detect respiratory complications, which can occur as the disease progresses and affects respiratory muscles. 7. Seizure Management – Seizure Control: Monitoring the frequency, severity, and response to antiepileptic medications to manage seizures effectively, which may occur in some cases of MLD. 8. Treatment Side Effects and Adherence – Medication Side Effects: Monitored through caregiver reports and clinical evaluations to ensure the safety and tolerability of treatments, such as enzyme replacement therapy or supportive care measures. – Adherence: Evaluated through caregiver reports and medication logs to ensure compliance with prescribed treatment regimens. 9. Social and Family Impact – Social Functioning: Assessed using tools like the Family Impact Scale (FIS) or the Pediatric Quality of Life Family Impact Module to evaluate the impact of MLD on family dynamics and social interactions. Consult with Our Team of Experts Now!
|
1.37 Pelizaeus-Merzbacher disease |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Neurological Function – Motor Function: Evaluated through clinical examinations and scales such as the Gross Motor Function Classification System (GMFCS) to assess muscle tone, strength, coordination, and motor skills. – Cognitive Function: Assessed using developmental scales appropriate for the patient’s age, such as the Bayley Scales of Infant and Toddler Development or specific cognitive assessment tools to evaluate intellectual abilities and developmental milestones. – Speech and Language: Evaluated through standardized tests and observations to assess communication skills, which may be affected due to neurological impairments. 2. Disease Progression – Neuroimaging: Utilized to monitor changes in the brain, particularly in the white matter, through techniques such as magnetic resonance imaging (MRI) to assess disease progression and structural abnormalities. – Electrophysiological Studies: Conducted to evaluate nerve conduction velocities and detect abnormalities in nerve function. 3. Functional Abilities – Activities of Daily Living (ADL): Assessed using scales like the Pediatric Evaluation of Disability Inventory (PEDI) or the Functional Independence Measure (FIM) to evaluate the patient’s ability to perform daily tasks independently. – Mobility and Gait: Evaluated using tests like the Timed Up and Go (TUG) test or the 6-Minute Walk Test (6MWT) to assess mobility, balance, and endurance. 4. Quality of Life – Health-Related Quality of Life (HRQoL): Measured using instruments such as the Pediatric Quality of Life Inventory (PedsQL) or disease-specific quality of life measures to assess the impact of PMD on the patient’s overall well-being and the family’s quality of life. – Emotional and Psychological Health: Assessed through caregiver reports and tools like the Pediatric Symptom Checklist (PSC) or specific behavioral assessment scales to evaluate emotional functioning and behavioral changes. 5. Genetic and Biomarker Assessments – Genetic Testing: Used to confirm the diagnosis of PMD by identifying mutations in genes associated with the disease, such as the PLP1 gene. – Biomarkers: Research is ongoing to identify biomarkers in blood or cerebrospinal fluid (CSF) that can help track disease progression and response to treatment. 6. Respiratory Function – Pulmonary Function Tests (PFTs): Regular spirometry to assess lung function and detect respiratory complications, which can occur as the disease progresses and affects respiratory muscles. 7. Seizure Management – Seizure Control: Monitoring the frequency, severity, and response to antiepileptic medications to manage seizures effectively, which may occur in some cases of PMD. 8. Treatment Side Effects and Adherence – Medication Side Effects: Monitored through caregiver reports and clinical evaluations to ensure the safety and tolerability of treatments, such as supportive care measures or symptom management. – Adherence: Evaluated through caregiver reports and medication logs to ensure compliance with prescribed treatment regimens. 9. Social and Family Impact – Social Functioning: Assessed using tools like the Family Impact Scale (FIS) or the Pediatric Quality of Life Family Impact Module to evaluate the impact of PMD on family dynamics and social interactions. Consult with Our Team of Experts Now!
|
1.38 Neurodegenerative disorders associated with aging (e.g., age-related cognitive decline, age-related macular degeneration) |
Adult-NSCs,BM‐MSCs,hUCB‐MSCs, DPSCs,ESCs, iPSCs Neuron-PSCs, Astro-PSCs, Oligo-PSCs, Micro-PSCs, Ependy-PSCs |
1. Cognitive Function – Global Cognitive Function: Assessed using tools like the Mini-Mental State Examination (MMSE) or the Montreal Cognitive Assessment (MoCA) to evaluate overall cognitive abilities. – Memory: Evaluated using specific tests like the Wechsler Memory Scale (WMS) or the California Verbal Learning Test (CVLT). – Executive Function: Assessed through tools like the Trail Making Test (TMT) or the Stroop Test. – Attention: Evaluated using the Digit Span test or the Continuous Performance Test (CPT). 2. Functional Outcomes – Activities of Daily Living (ADL): Assessed using scales like the Barthel Index or the Functional Independence Measure (FIM) to evaluate the patient’s ability to perform daily tasks independently. – Instrumental Activities of Daily Living (IADL): Evaluated through tools like the Lawton IADL Scale to assess more complex activities necessary for independent living, such as managing finances or medication. 3. Quality of Life – Health-Related Quality of Life (HRQoL): Measured using instruments like the SF-36 Health Survey or the Quality of Life in Alzheimer’s Disease (QoL-AD) scale. – Emotional and Psychological Health: Assessed using tools like the Geriatric Depression Scale (GDS) or the Patient Health Questionnaire-9 (PHQ-9) for depression and the Generalized Anxiety Disorder 7 (GAD-7) scale for anxiety. 4. Visual Function (Specific to Age-Related Macular Degeneration) – Visual Acuity: Assessed using the Snellen chart or the Early Treatment Diabetic Retinopathy Study (ETDRS) chart to measure central vision sharpness. – Contrast Sensitivity: Evaluated using tests like the Pelli-Robson chart to assess the ability to distinguish between different shades of grey. – Visual Field Testing: Conducted using perimetry tests to evaluate peripheral vision. – Retinal Imaging: Regularly performed using optical coherence tomography (OCT) and fundus photography to monitor structural changes in the retina. 5. Disease Progression – Neuroimaging: Regular MRI or CT scans to monitor brain structure and detect atrophy in neurodegenerative conditions like Alzheimer’s disease. – Biomarkers: Research is ongoing to identify biomarkers in cerebrospinal fluid (CSF) or blood that can help track disease progression and response to treatment. 6. Physical Function – Mobility and Balance: Assessed using the Timed Up and Go (TUG) test or the Berg Balance Scale to evaluate the risk of falls and mobility limitations. – Strength and Endurance: Evaluated through tests like the 6-Minute Walk Test (6MWT) or grip strength measurements. 7. Sleep and Behavior – Sleep Quality: Assessed using tools like the Pittsburgh Sleep Quality Index (PSQI) to evaluate sleep disturbances. – Behavioral Changes: Monitored through clinical interviews and behavioral assessments to detect changes in mood, agitation, or aggression. 8. Medication Side Effects and Adherence – Side Effects: Monitored through patient self-reports and clinical evaluations to ensure the safety and tolerability of treatments. – Adherence: Evaluated through patient self-reports, pill counts, or electronic monitoring to ensure compliance with prescribed treatment regimens. 9. Social Functioning – Social Engagement: Assessed using tools like the Lubben Social Network Scale (LSNS) to evaluate social connections and support. – Caregiver Burden: Evaluated using tools like the Zarit Burden Interview (ZBI) to assess the impact of caregiving on family members. 10. Pain and Comfort – Pain Assessment: Tools like the Numeric Pain Rating Scale (NPRS) or the Brief Pain Inventory (BPI) are used to assess pain levels and discomfort. – Comfort Measures: Evaluated through patient and caregiver reports to ensure optimal comfort and quality of life. Consult with Our Team of Experts Now!
|
63. ^ Alzheimer’s Disease and Cognitive Function Assessment
64. Amyotrophic Lateral Sclerosis (ALS) Functional Rating Scale
65. Assessment Tools for Autism Spectrum Disorder
66. Scale for the Assessment and Rating of Ataxia (SARA)
67. Cerebral Palsy and Motor Function Assessment
68. Quality of Life in Patients with Epilepsy
69. Huntington’s Disease Rating Scale
70. ^ Expanded Disability Status Scale (EDSS) in Multiple Sclerosis