Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) represent a transformative frontier in the field of regenerative neurology. ALS, also known as Lou Gehrig’s disease, is a relentlessly progressive neurodegenerative disorder that selectively targets motor neurons in the brain and spinal cord. This degeneration leads to muscle wasting, paralysis, respiratory failure, and ultimately death, typically within 3 to 5 years of diagnosis. Traditional treatment strategies like riluzole and edaravone offer only modest symptomatic relief and minimal impact on disease progression. In this context, the regenerative approach offered by Cellular Therapy and Stem Cells provides a bold, innovative avenue toward neuroprotection, neuroregeneration, and functional restoration.
Our focus is to explore how regenerative medicine can shift the paradigm from disease management to meaningful repair in ALS. Through a combination of mesenchymal stem cells (MSCs), neural stem cells (NSCs), induced pluripotent stem cells (iPSCs), and exosome-based therapies, the aim is to restore failing motor neuron circuits, modulate neuroinflammation, and slow the irreversible neurodegenerative process at its core. From localized stem cell implantation to intravenous, intrathecal, and intranasal delivery routes, a range of personalized protocols are actively being developed at our center, reflecting the highest standard of ethical sourcing, advanced cellular manipulation, and scientific rigor.
Let us walk you through this revolution—where hope meets science, and innovation meets action—for patients confronting the devastating reality of ALS [1-5].
Before administering Cellular Therapy and Stem Cells for ALS, our center emphasizes the importance of personalized genomic profiling. ALS has both sporadic and familial forms. While the sporadic type accounts for the majority of cases, up to 10% of patients have familial ALS linked to inherited gene mutations. Genetic testing helps illuminate individual risk factors, disease trajectory, and therapeutic responsiveness.
Our team utilizes cutting-edge DNA sequencing platforms to detect mutations in key genes associated with ALS, such as:
By identifying these genetic vulnerabilities, our clinicians can develop personalized stem cell treatment protocols—tailoring cell type, delivery route, and adjunctive therapies to a patient’s unique genomic profile. This integrative approach maximizes therapeutic potential while minimizing unnecessary interventions. Early risk detection also opens the door to preventive lifestyle interventions, targeted neuroprotective agents, and proactive family screening [1-5].
ALS is a devastating neurological disorder marked by the progressive degeneration of upper and lower motor neurons, leading to paralysis and premature death. The pathogenesis is multifactorial, involving a complex interplay of genetic, molecular, cellular, and inflammatory mechanisms. Understanding these underpinnings is essential for designing effective Cellular Therapy and Stem Cell interventions.
Oxidative Stress and Mitochondrial Dysfunction
Motor neurons are highly susceptible to oxidative stress due to their long axons and high metabolic demand. In ALS, defective mitochondria result in ATP depletion, increased reactive oxygen species (ROS), and neuronal apoptosis.
Excitotoxicity
Dysregulation of the glutamate transporter EAAT2 in astrocytes causes extracellular glutamate accumulation, overstimulating neurons and leading to calcium overload and excitotoxic death.
Protein Aggregation
Mutations in genes like SOD1, TDP-43, and FUS cause misfolded proteins to accumulate in motor neurons. These aggregates disrupt axonal transport, RNA processing, and cellular homeostasis.
Microglial Activation
Microglia, the immune cells of the CNS, become hyperactivated in ALS, releasing pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) and nitric oxide. This neuroinflammatory environment accelerates neuronal degeneration.
Astrocyte Toxicity
Mutant astrocytes lose their ability to support motor neurons and instead release toxic mediators that contribute to motor neuron death.
The cytoskeleton-dependent transport of essential proteins and organelles along axons is disrupted in ALS. Mutations in dynein and other motor proteins impair bidirectional transport, leading to neuronal dysfunction.
Early in ALS, synaptic inputs to motor neurons are lost—termed “dying-back” degeneration—before the neurons themselves die. This synaptic breakdown contributes to early muscle weakness.
As motor neurons degenerate, muscles become denervated, leading to progressive atrophy, fasciculations, and spasticity. Diaphragmatic involvement ultimately causes respiratory failure.
ALS also affects autonomic and cognitive functions in some patients, especially in C9orf72 mutation carriers. These patients may develop frontotemporal dementia (FTD) alongside classical ALS symptoms [1-5].
The core aim of Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) is not just neuroprotection, but neurorestoration. Multiple strategies are currently in clinical development or compassionate use at DrStemCellsThailand’s Center, including:
The effectiveness of Cellular Therapy often hinges on precise delivery. We employ:
Each route is selected based on disease stage, anatomical progression, and genetic background—ensuring a tailored, effective treatment strategy.
ALS has long been considered an untreatable condition. But today, at the intersection of regenerative medicine, neurogenetics, and immunology, that narrative is beginning to change. Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) offer more than a glimmer of hope—they offer a roadmap to rewire the disease course. Our commitment at DrStemCellsThailand is not just to extend life, but to restore meaning to it—by giving patients renewed function, dignity, and time [1-5].
Amyotrophic Lateral Sclerosis (ALS), also known as Lou Gehrig’s disease, is a relentlessly progressive neurodegenerative disorder that affects upper and lower motor neurons. It leads to profound muscle atrophy, paralysis, and ultimately respiratory failure. The pathogenesis of ALS is multifactorial, involving a synergistic blend of genetic mutations, cellular dysfunction, environmental exposures, and inflammatory responses.
Neuroinflammation and Oxidative Stress
One of the earliest and most persistent hallmarks of ALS is neuroinflammation accompanied by oxidative damage. Activated microglia and astrocytes in the central nervous system release high levels of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) and nitric oxide, resulting in the progressive destruction of motor neurons.
Simultaneously, the buildup of reactive oxygen species (ROS) and reactive nitrogen species (RNS) due to impaired mitochondrial function exacerbates oxidative stress, directly injuring DNA, lipids, and neuronal cytoskeleton.
Glutamate Excitotoxicity
In ALS, there is impaired reuptake of glutamate by astrocytes due to dysfunction of the EAAT2 transporter. This causes excessive extracellular glutamate, overstimulating NMDA and AMPA receptors on motor neurons.
Prolonged activation of these receptors leads to intracellular calcium overload, mitochondrial dysfunction, and neuronal apoptosis.
Mitochondrial Dysfunction
Mitochondria in ALS-affected neurons exhibit abnormal morphology, impaired ATP production, and increased permeability.
Mitochondrial DNA mutations, reduced oxidative phosphorylation, and cytochrome c release play a crucial role in triggering caspase activation and apoptosis in degenerating motor neurons.
Protein Aggregation and Impaired Autophagy
ALS is characterized by the abnormal accumulation of misfolded proteins such as TDP-43, SOD1, and FUS within motor neurons.
These protein aggregates overwhelm the proteasome system and disrupt autophagy, leading to toxic intracellular inclusions that damage organelles and impair synaptic communication [6-10].
Genetic Mutations and Familial ALS
Around 10% of ALS cases are familial, linked to mutations in genes such as:
These mutations initiate neurodegeneration through multiple converging mechanisms, including mitochondrial damage, axonal transport dysfunction, and abnormal RNA splicing.
Environmental and Epigenetic Triggers
Environmental factors such as pesticide exposure, smoking, heavy metals, and viral infections may increase ALS risk by initiating neuroinflammation and genotoxic stress.
Epigenetic alterations including DNA methylation, histone modification, and non-coding RNA dysregulation also play critical roles in altering gene expression in vulnerable motor neuron populations.
Given the heterogeneous and multifactorial nature of ALS, a single-target approach is insufficient. Multimodal, regenerative strategies such as Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) offer new hope in addressing these complex mechanisms [6-10].
Despite decades of research, treatment for ALS remains largely supportive. Current pharmacologic agents only offer modest survival benefits, and the underlying neurodegeneration continues unchecked. Key limitations include:
Lack of Curative Therapies
FDA-approved drugs like riluzole and edaravone may slow disease progression but fail to prevent motor neuron loss or restore lost function.
No conventional treatment has been shown to regenerate damaged neurons or reverse muscle wasting.
Rapid and Irreversible Neurodegeneration
ALS progresses rapidly, with most patients losing the ability to walk, speak, or breathe unassisted within 2 to 5 years of diagnosis.
Traditional therapies cannot keep pace with the aggressive nature of neurodegeneration.
Ineffective in Modulating Neuroinflammation and Excitotoxicity
Available treatments do not sufficiently reduce glutamate excitotoxicity, oxidative damage, or inflammatory cascades that drive motor neuron death.
Barriers to Drug Delivery Across the Blood-Brain Barrier (BBB)
Many potentially neuroprotective compounds fail due to poor penetration across the BBB, making it difficult to reach affected motor neurons in the brain and spinal cord.
Lack of Personalized or Regenerative Approaches
Current therapies are not tailored to individual genetic or pathological profiles and offer no potential for neuronal repair or functional restoration.
These significant challenges highlight the urgent need for transformative strategies like Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS), capable of neuroprotection, neuroregeneration, and systemic immune modulation [6-10].
The integration of regenerative medicine into ALS treatment is rapidly evolving. Stem cell-based therapies aim to replace lost neurons, modulate inflammation, restore neuromuscular communication, and delay disease progression. Groundbreaking innovations include:
Personalized Regenerative Protocols for ALS
Year: 2004
Researcher: Our Medical Team
Institution: DrStemCellsThailand‘s Anti-Aging and Regenerative Medicine Center of Thailand
Result: Our Medical Team’s pioneering therapeutic protocol for ALS employs autologous and allogeneic mesenchymal stem cells (MSCs), neural progenitor stem cells (NPCs), and exosome-enriched biologics. Administered intrathecally and intravenously, this regimen targets neuroinflammation, promotes neuronal survival, and improves muscular function in ALS patients globally.
Mesenchymal Stem Cell (MSC) Therapy
Year: 2012
Researcher: Dr. Eva Feldman
Institution: University of Michigan, USA
Result: Intrathecal delivery of bone marrow-derived MSCs in ALS patients was shown to reduce pro-inflammatory cytokine levels and preserve motor function, with some patients demonstrating improved respiratory capacity.
Neural Stem Cell (NSC) Therapy
Year: 2016
Researcher: Dr. Nicholas Boulis
Institution: Emory University School of Medicine, USA
Result: Human spinal cord-derived NSCs were surgically transplanted into the lumbar spinal cord of ALS patients, leading to stabilization of motor symptoms in early-phase clinical trials [6-10].
Induced Pluripotent Stem Cell (iPSC)-Derived Motor Neurons
Year: 2018
Researcher: Dr. Shinya Yamanaka
Institution: Kyoto University, Japan
Result: Patient-specific iPSCs were differentiated into motor neurons and used to model ALS in vitro, allowing for individualized drug testing and personalized therapeutic design. Transplantation studies in animal models have shown partial restoration of neuromuscular function.
Extracellular Vesicles (EVs) from MSCs
Year: 2020
Researcher: Dr. Magdalena Gołąbek
Institution: Institute of Experimental Biology, Poland
Result: MSC-derived EVs reduced glial activation, inhibited neurotoxic cytokine release, and preserved neuromuscular junction integrity in ALS mouse models.
Combination Therapy with Exosomes, Growth Factors, and Peptides
Year: 2023
Researcher: Dr. Lars Ljungberg
Institution: Lund University, Sweden
Result: A cocktail of exosomes, BDNF, GDNF, and neuropeptides derived from umbilical MSCs demonstrated synergistic effects in rescuing motor neurons and improving motor coordination in ALS models.
These pioneering therapies underscore the regenerative potential of stem cells and associated biologics in redefining ALS management [6-10].
ALS has gained widespread attention due to its devastating impact and the involvement of influential figures who champion awareness and research for a cure:
Stephen Hawking
The renowned physicist defied the odds by living with ALS for over five decades. His global presence brought ALS into the spotlight and inspired generations to support research into its mechanisms and treatment.
Steve Gleason
The former NFL player founded Team Gleason, a nonprofit focused on improving the lives of ALS patients through assistive technology and regenerative research.
Lou Gehrig
The legendary baseball player for whom the disease was named. His farewell speech and public struggle with ALS forever linked the condition to courage and the need for medical advancement.
O. J. Brigance
Former NFL linebacker and Super Bowl champion who founded the Brigance Brigade Foundation to raise awareness and funding for ALS treatment innovations.
Augie Nieto
The fitness industry icon, diagnosed with ALS in 2005, has helped raise millions of dollars for regenerative ALS research through the ALS Therapy Development Institute.
These individuals have elevated public consciousness of ALS and underscored the importance of pursuing regenerative medicine solutions like Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) [6-10].
Preventing the progression of pancreatic neuroendocrine tumors (PNETs) requires early, proactive, and biologically targeted intervention. Our comprehensive immunotherapy protocols merge tumor-specific immunologic precision with regenerative potential:
By targeting the neuroendocrine tumor microenvironment and immune escape mechanisms, our Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) provide a forward-thinking, precision-guided strategy to halt tumor progression and restore immune surveillance [11-15].
Time-sensitive intervention is essential in managing PNETs, which often present indolently but can rapidly become invasive and metastatic. Early application of cellular immunotherapy profoundly alters disease trajectory:
Patients receiving immunotherapy during early tumor stages exhibit higher remission rates, reduced endocrine disruption, and better preservation of exocrine pancreatic function. Timely enrollment in our Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) program significantly boosts tumor control, endocrine balance, and long-term survival [11-15].
PNETs present a unique challenge due to their immunologically “cold” nature and endocrine functionality. Our therapies are engineered to overcome these defenses through cellular mechanisms rooted in immune engineering and tumor biology:
These targeted properties make Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) a mechanistically robust and adaptable platform for overcoming tumor resistance and enabling durable disease control [11-15].
PNETs progress through a clinically recognized spectrum, from low-grade tumors to metastatic carcinomas. Recognizing each stage’s biological profile allows for tailored cellular immunotherapeutic interventions:
Stage 1: Localized Low-Grade PNET
Stage 2: Locally Invasive PNET
Stage 3: Lymph Node Metastasis
Stage 4: Hepatic Metastasis
Stage 5: Endocrine Crisis or Functional Tumor Syndrome
Each phase of PNET evolution benefits from a tailored cellular immunotherapy strategy, balancing tumor control with endocrine stability [11-15].
Stage 1: Localized PNET
Stage 2: Invasive PNET
Stage 3: Lymphatic Spread
Stage 4: Liver Metastasis
Stage 5: Endocrine Crisis
This layered approach integrates cellular immunotherapy seamlessly into standard-of-care PNET management, enhancing efficacy at each disease stage [11-15].
Our Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) program for Pancreatic Neuroendocrine Tumors redefines conventional boundaries of oncology and immunology:
This approach offers a transformative vision for PNET therapy—one where tumors are dismantled, endocrine functions are preserved, and immune control becomes durable and dynamic [11-15].
Allogeneic Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) offers an efficient, potent, and scalable option for modern cancer immunotherapy, especially in complex or refractory neuroendocrine cases [11-15].
Preventing neurodegenerative progression in ALS requires early regenerative interventions targeting motor neuron survival and neuroinflammation. Our Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) protocol strategically incorporates:
Through these regenerative modalities, we offer a cutting-edge neurorestorative framework designed to halt ALS progression and promote functional recovery [16-20].
Our neurology and regenerative medicine experts emphasize the importance of early therapeutic intervention in ALS to preserve upper and lower motor neurons before irreversible degeneration sets in:
Early enrollment in our Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) program offers the best chance to preserve neuromuscular integrity and delay disease escalation [16-20].
ALS is marked by progressive motor neuron degeneration, glial activation, and neuroinflammation. Our regenerative strategy leverages stem cell-based interventions to directly address ALS’s multifactorial pathophysiology:
These cellular approaches allow us to counteract the neurodegenerative spiral of ALS at its roots—structural, biochemical, and immunologic [16-20].
ALS progression follows a clinically recognized pattern of motor and respiratory decline. Regenerative interventions must be tailored to these evolving stages:
Our comprehensive ALS program integrates:
By disrupting ALS’s neurodegenerative cascade and promoting neural repair, we offer hope for improved outcomes beyond conventional pharmacology [16-20].
Through advanced allogeneic Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS), our program aims to optimize safety, consistency, and therapeutic potency for all patients [16-20].
Our allogeneic Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) harnesses the regenerative and neuroprotective power of ethically sourced, high-potency stem cells. These cells target motor neuron degeneration and neuroinflammation—key pathological hallmarks of ALS progression:
Umbilical Cord-Derived MSCs (UC-MSCs): UC-MSCs exhibit robust immunomodulatory activity and secrete neurotrophic factors such as brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF), which support motor neuron survival and mitigate microglial activation.
Wharton’s Jelly-Derived MSCs (WJ-MSCs): Known for their superior proliferative capacity, WJ-MSCs reduce oxidative stress and promote remyelination by releasing extracellular vesicles enriched with miRNAs and exosomes that foster motor neuron repair.
Placenta-Derived Stem Cells (PLSCs): These cells secrete hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF), enhancing neural tissue perfusion and protecting against ischemic neuronal injury in ALS.
Amniotic Fluid Stem Cells (AFSCs): AFSCs are uniquely suited for neuroregeneration, as they can differentiate into neural lineages and release anti-apoptotic factors that limit neuronal loss in the spinal cord.
Neural Progenitor Cells (NPCs): NPCs directly integrate into degenerative motor neuron niches, where they differentiate into astrocytes and oligodendrocytes, restoring glial support and modulating excitotoxic environments in ALS patients.
Our multi-source cellular matrix enhances synaptic connectivity, reduces inflammatory cytokines (IL-1β, TNF-α), and maximizes neuroplasticity, offering a promising therapeutic avenue for ALS [21-23].
Our regenerative medicine laboratory is committed to delivering the safest and most scientifically validated stem cell treatments for ALS:
Regulatory Compliance and Certification: Fully licensed by the Thai FDA, our protocols comply with Good Manufacturing Practice (GMP) and Good Laboratory Practice (GLP) standards, ensuring reproducibility and safety.
Advanced Cleanroom Infrastructure: All stem cell processing is conducted in ISO4/Class 10 cleanroom environments, reducing contamination risks and preserving cellular viability.
Preclinical and Clinical Validation: Supported by translational research and preclinical ALS models that confirm neuroprotective efficacy, with ongoing clinical trials shaping protocol refinements.
Customized Protocol Development: Each patient’s ALS stage, progression rate, and biomarker profile guide the selection of cell type, dosage, and delivery method—typically intrathecal and intravenous.
Ethical, Non-Invasive Sourcing: All allogeneic cells are derived from ethically approved tissue donations, ensuring sustainability and respect for human life.
This integrated model of quality control and scientific rigor guarantees the highest standards in Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) [21-23].
To assess treatment effectiveness in ALS patients, we track motor function scores (ALSFRS-R), electromyography (EMG) signals, respiratory capacity (FVC), and neuroinflammatory biomarkers. Our Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) have demonstrated:
Preservation of Motor Neurons: MSC-derived neurotrophic factors protect upper and lower motor neurons, slowing denervation and axonal degradation.
Neuroinflammatory Suppression: Intrathecal stem cell administration significantly reduces CSF concentrations of IL-6, MCP-1, and TNF-α.
Enhanced Synaptic Plasticity: Neural progenitor cells promote synaptogenesis and remyelination, aiding signal conduction and motor strength.
Improved Quality of Life: Patients report better mobility, slower decline in respiratory function, and improved emotional well-being due to reduced neurological burden.
By addressing both inflammation and neurodegeneration, our regenerative approach offers an innovative alternative to symptom management, with the potential to decelerate ALS progression [21-23].
Our regenerative specialists evaluate each ALS patient thoroughly to determine suitability for our advanced cellular therapies. Not all individuals with ALS may qualify due to the systemic burden and progression of the disease.
Exclusion Criteria:
Pre-Treatment Optimization:
Strict eligibility safeguards ensure optimal clinical outcomes and reduce adverse events [21-23].
Some patients with moderately advanced ALS may still benefit from our protocols, particularly those with preserved respiratory function and stable comorbidities. Clinical review includes:
Neuroimaging: Cervical and thoracic spinal MRI to evaluate motor neuron atrophy and corticospinal tract degeneration.
Functional Assessments: ALSFRS-R score tracking over 3 months, FVC percentage, and limb strength measurements.
CSF Biomarkers: Levels of neurofilament light chains (NfL), glial fibrillary acidic protein (GFAP), and cytokines.
Genetic and Immune Screening: Identification of SOD1, TDP-43, or FUS mutations and autoimmune antibodies.
Respiratory and Nutritional Review: Spirometry and body composition analysis.
These evaluations allow us to determine whether cellular therapy may decelerate ALS progression, particularly in younger patients with slower disease trajectories [21-23].
Our international qualification process ensures each ALS patient receives safe and personalized care. The multidisciplinary team performs a complete review of:
Recent test results (within 3 months) are mandatory. These evaluations help us customize and calibrate stem cell therapy for ALS on a patient-specific basis [21-23].
Once qualified, each patient receives a comprehensive treatment plan including:
Cell Type and Dosage: Administration of 80–150 million MSCs and NPCs derived from umbilical cord, Wharton’s Jelly, amniotic fluid, and placental sources.
Delivery Routes:
Follow-up: Structured evaluations over 3, 6, and 12 months to assess functional stabilization [21-23].
The therapeutic process spans 10–14 days in Thailand and includes:
Multimodal Stem Cell Delivery:
Supportive Regenerative Therapies:
Cost Overview:
Our Cellular Therapy and Stem Cells for Amyotrophic Lateral Sclerosis (ALS) program is an innovative alternative for patients seeking functional preservation and quality of life improvement beyond conventional interventions [21-23].