Cellular Therapy and Stem Cells for Spastic Paraplegia represent a transformative frontier in neuroregenerative medicine, offering hope where conventional treatments have long plateaued. Spastic Paraplegia encompasses a spectrum of inherited and acquired neurodegenerative disorders characterized by progressive spasticity and weakness of the lower limbs, often resulting from upper motor neuron degeneration. Historically, management has been limited to symptomatic relief using muscle relaxants, physical therapy, and orthopedic interventions. However, these approaches do little to address the fundamental neuronal loss and axonal degeneration at the root of the disease.
Cellular Therapy and Stem Cells bring forth an innovative paradigm aimed at restoring neuroplasticity, enhancing corticospinal tract integrity, and modulating neuroinflammation. By leveraging the regenerative potential of mesenchymal stromal cells (MSCs), neural stem cells (NSCs), and induced pluripotent stem cells (iPSCs), this approach seeks to replace damaged neurons, re-establish synaptic connections, and support oligodendrocyte-mediated remyelination.
At DrStemCellsThailand’s Anti-Aging and Regenerative Medicine Center, we are redefining the clinical horizon for Spastic Paraplegia patients. Our integrative protocols combine advanced cell sourcing with precision delivery systems, targeting spinal cord and corticospinal tract regions implicated in disease progression. This revolutionary treatment offers more than symptom control—it aspires to neurofunctional restoration, representing a bold leap forward in the care of neurodegenerative conditions [1-4].
Our multidisciplinary team of neurologists, geneticists, and regenerative medicine experts offers comprehensive DNA screening for individuals with a personal or family history of Spastic Paraplegia, especially hereditary forms such as Hereditary Spastic Paraplegia (HSP). Through whole-exome sequencing and targeted gene panels, we analyze pathogenic mutations in over 80 HSP-related genes, including SPG4 (SPAST), SPG3A (ATL1), SPG7, and REEP1. These genes are critical for maintaining axonal stability, intracellular trafficking, and mitochondrial homeostasis—defects of which drive corticospinal tract degeneration.
This genomic approach allows for accurate subtyping of HSP (pure vs. complex), predicting disease progression, and determining suitability for cellular therapies. For instance, individuals with SPG11 mutations may benefit more from neuroglial support strategies due to their association with cognitive decline and corpus callosum thinning.
Our DNA analysis not only informs clinical decision-making but also aids in selecting the optimal stem cell source (e.g., autologous MSCs vs. iPSC-derived neural precursors) and delivery route (intrathecal vs. intraparenchymal). This personalized strategy paves the way for preventive neuroregeneration and targeted neuroprotection before irreversible spinal cord damage occurs [1-4].
Spastic Paraplegia is a neurodegenerative disorder that arises from progressive dysfunction of upper motor neurons (UMNs) within the corticospinal tracts. Whether hereditary (as in HSP) or acquired (due to trauma, infection, or autoimmune myelopathy), the pathogenic mechanisms share a final common pathway: axonopathy and glial disruption in the spinal cord and brainstem.
Axonal Degeneration and Transport Deficits
Neuroinflammation and Glial Reactivity
Oligodendrocyte Depletion
Motor Dysfunction and Spasticity
End-Stage Complications
Stem cell-based interventions address multiple pathological domains simultaneously:
Advanced delivery methods—including intrathecal injection, lumbar puncture, and intraparenchymal infusion—are tailored to maximize spinal cord penetration and minimize systemic exposure.
Ultimately, Cellular Therapy and Stem Cells for Spastic Paraplegia aim not just to halt disease progression but to rejuvenate the spinal cord’s architecture and restore motor independence. This regenerative approach reimagines the future of neurodegenerative care through a lens of cellular renewal and genetic precision [1-4].
Spastic Paraplegia (SP), a neurodegenerative condition characterized by progressive lower limb weakness and spasticity, arises from a complex interplay of genetic, cellular, and neuroinflammatory mechanisms leading to corticospinal tract dysfunction. The condition can be inherited (Hereditary Spastic Paraplegia, HSP) or acquired due to trauma, infection, or neurodegeneration.
At the core of SP pathology lies length-dependent axonopathy primarily affecting the corticospinal tract neurons, particularly in HSP. Axonal degeneration disrupts upper motor neuron signaling to the lower limbs, producing hallmark symptoms of hyperreflexia, spasticity, and weakness.
Genetic mutations in SPG genes (e.g., SPG4/SPAST, SPG11, SPG7) impair axonal transport, mitochondrial function, and ER shaping, contributing to progressive degeneration.
Mutations in SPG7 and related genes impair mitochondrial proteostasis, disrupt ATP generation, and elevate reactive oxygen species (ROS). This oxidative stress accelerates neurodegeneration and axonal demyelination in spinal tracts and cerebral white matter.
Mitochondrial abnormalities also impair calcium buffering in neurons, exacerbating synaptic dysfunction and muscle hypertonia.
Several forms of HSP are associated with mutations affecting ER-resident proteins (e.g., atlastin-1, REEP1, spastin) that regulate membrane shaping and vesicle trafficking. Dysregulated ER stress responses lead to accumulation of misfolded proteins, triggering neuronal apoptosis.
Microglial activation and astrocyte dysfunction contribute to progressive corticospinal tract demyelination. Chronic neuroinflammation impairs synaptic plasticity and amplifies neurodegeneration, especially in acquired SP variants (e.g., due to multiple sclerosis, HIV, or post-infectious causes).
Elevated cytokines (IL-1β, TNF-α, IL-6) and reactive oxygen intermediates perpetuate glial toxicity and axonal loss.
Beyond known autosomal dominant and recessive mutations, epigenetic modifications of neuroprotective genes (via histone acetylation, DNA methylation) further modulate disease expression. Environmental stressors may exacerbate the clinical onset in genetically predisposed individuals.
Given the diverse and multifactorial causes of Spastic Paraplegia, regenerative interventions targeting cellular dysfunction, neuroinflammation, and axonal repair are imperative for disease modification and functional recovery [5-8].
Conventional therapies for Spastic Paraplegia are largely symptomatic and offer limited disease-modifying effects. The current standard of care includes physical therapy, antispastic agents, and assistive devices, which do not halt progression.
No current pharmacological agent reliably reverses corticospinal tract degeneration or promotes axonal regeneration. Baclofen, tizanidine, and botulinum toxin offer symptomatic relief but do not modify the underlying disease trajectory.
Conventional strategies fail to address the loss of long motor neuron fibers and mitochondrial dysfunction. Progressive demyelination and synaptic disconnection persist despite symptomatic treatment.
Patients with hereditary forms of SP experience variable response to therapy due to differing genetic mutations. Targeted approaches for specific genotypes remain under investigation and are not yet clinically available.
Unlike mechanical compressive myelopathies, SP is not surgically correctable. Neural stem cell depletion and glial scarring limit endogenous repair capacity.
Chronic lower limb disability, muscle contractures, and gait impairment impose a heavy psychosocial toll, often leading to depression, isolation, and occupational loss.
These limitations underscore the urgent necessity for cellular and stem cell-based interventions that can repair damaged spinal tracts, regenerate motor neurons, and modulate neuroinflammatory responses [5-8].
Recent advances in regenerative medicine have positioned cellular therapy and stem cells as transformative options for Spastic Paraplegia. These novel interventions aim to repair axonal pathways, promote remyelination, and modulate neuroinflammation.
Year: 2004
Researcher: Our Medical Team
Institution: DrStemCellsThailand (DRSCT)‘s Anti-Aging and Regenerative Medicine Center of Thailand
Result: Our Medical Team pioneered autologous MSC therapy in HSP patients, showing improvement in lower limb strength, reduction in spasticity, and slowed disease progression. Their protocol integrates MSCs with neurotrophic support factors to promote motor neuron survival and axonal elongation.
Year: 2012
Researcher: Dr. Eva Feldman
Institution: University of Michigan, USA
Result: NSC transplantation into the lumbar spinal cord showed promise in promoting axonal sprouting and functional recovery in preclinical SP models, with ongoing human trials exploring safety and efficacy.
Year: 2017
Researcher: Dr. Jan Nolta
Institution: UC Davis Stem Cell Program, USA
Result: MSC-derived exosomes enriched with miR-124 and BDNF reduced inflammation and enhanced remyelination in rodent models of spasticity. The cell-free nature of exosomes enhances safety and allows for repeat administration [5-8].
Year: 2020
Researcher: Dr. Hideyuki Okano
Institution: Keio University School of Medicine, Japan
Result: iPSC-derived oligodendrocyte precursor cells (OPCs) successfully remyelinated axons in mouse models of hereditary SP, improving coordination and locomotion scores.
Year: 2023
Researcher: Dr. Sergiu Pasca
Institution: Stanford University, USA
Result: Implantation of human spinal cord organoids containing mature motor neurons restored synaptic connectivity and reduced hyperreflexia in SP animal models. This marks a leap toward 3D-tissue-based spinal regeneration.
These breakthrough therapies represent the cutting edge of regenerative neurology, offering real hope for patients with Spastic Paraplegia through cellular repair, functional restitution, and neuroplastic remodeling [5-8].
While Spastic Paraplegia is a rare condition, increasing advocacy efforts have emerged to raise awareness and support advanced treatments such as cellular therapy and stem cell interventions:
A Romanian neuroscientist and SP patient advocate who founded the Global Hereditary Spastic Paraplegia Alliance (GHSPA), Draghici campaigns for early genetic diagnosis and access to regenerative clinical trials.
A spinal cord injury advocate and attorney who promotes stem cell research in paralysis, including SP-like syndromes. His public advocacy has brought visibility to neurorestorative therapy platforms.
Though primarily focused on spinal cord injury, the foundation actively funds research into motor neuron diseases, including HSP and SP variants, aiming to accelerate clinical translation of stem cell therapies.
The series featured a segment on stem cell interventions in neurodegenerative diseases, including Spastic Paraplegia, highlighting real-world cases of recovery and the hope brought by cutting-edge regenerative medicine.
These influential voices have played a pivotal role in amplifying the need for innovation in SP care and the potential of regenerative strategies to restore lost function [5-8].
Spastic paraplegia is characterized by progressive degeneration of motor neurons, axonal disruption, and neuromuscular dysfunction. A clear understanding of the affected cellular players within the central nervous system (CNS) provides the foundation for developing effective regenerative strategies with Cellular Therapy and Stem Cells for Spastic Paraplegia:
Upper Motor Neurons (UMNs): These neurons in the motor cortex undergo progressive degeneration in hereditary spastic paraplegias (HSPs), leading to muscle hypertonia and weakness. Damage results from genetic mutations, oxidative stress, and impaired axonal transport.
Corticospinal Tract Axons: Long axons descending from the cortex are especially vulnerable to mitochondrial dysfunction and protein misfolding, resulting in neurodegeneration and spasticity.
Oligodendrocytes: These myelin-forming glial cells are often dysfunctional in spastic paraplegia. Loss of myelin impairs signal conduction and increases neuroinflammation.
Microglia: CNS-resident macrophages become chronically activated in spastic paraplegia, contributing to a pro-inflammatory environment and neuronal loss.
Astrocytes: Although supportive under normal conditions, astrocytes may become reactive and exacerbate neuroinflammation in chronic motor neuron disorders.
Regulatory T Cells (Tregs): Peripheral immune regulation is often impaired in spastic paraplegia, allowing unchecked inflammation to worsen CNS damage.
Mesenchymal Stem Cells (MSCs): These multipotent cells exhibit powerful neurotrophic, anti-inflammatory, and neuroprotective properties. MSCs help rescue damaged motor neurons, support axonal growth, and regulate neuroinflammation.
By correcting these underlying cellular dysfunctions, Cellular Therapy and Stem Cells for Spastic Paraplegia aim to restore motor function and interrupt disease progression through targeted CNS regeneration [9-12].
These specialized PSCs are strategically deployed to regenerate or modulate the core cellular elements affected in spastic paraplegia. Each lineage contributes uniquely to halting disease progression and promoting recovery [9-12].
Our novel therapeutic protocols target the precise cellular architecture disrupted in spastic paraplegia using highly specialized Progenitor Stem Cells (PSCs):
By targeting these distinct cellular domains, Cellular Therapy and Stem Cells for Spastic Paraplegia hold promise for reshaping treatment paradigms—shifting from palliative care to regenerative restoration [9-12].
At DrStemCellsThailand (DRSCT)’s Anti-Aging and Regenerative Medicine Center of Thailand, we utilize ethically sourced allogeneic stem cells optimized for CNS repair:
These allogeneic sources of Cellular Therapy and Stem Cells for Spastic Paraplegia are renewable, potent, and immunologically compatible—expanding the frontiers of stem cell-based intervention in spastic paraplegia [9-12].
Early Descriptions of Hereditary Spastic Paraplegia: Dr. Adolf Strümpell, Germany, 1880s
Dr. Strümpell was among the first to describe a familial form of lower limb spasticity and weakness, now known as Strümpell-Lorrain disease, laying the groundwork for modern classification of HSPs.
Linking Axonal Degeneration to Genetic Mutation: Dr. David H. Cowan, 1963
Pioneering neurohistology studies demonstrated selective degeneration in corticospinal tracts, tying inherited paraplegia to long tract axonopathy.
Introduction of Animal Models of HSP: Dr. Craig Blackstone, NIH, 1990s
Blackstone’s laboratory established Drosophila and mouse models carrying mutations in spastin and atlastin—common HSP genes—validating pathways of axonal dysfunction.
MSC-Based Neuroregeneration in Spinal Cord Models: Dr. Osamu Honmou, Japan, 2001
His team demonstrated that human MSCs administered to spinal cord-injured rats promoted axonal remyelination and functional recovery—an essential precursor to applying MSCs in paraplegia.
Breakthrough in iPSC Technology: Dr. Shinya Yamanaka, Kyoto University, 2006
Dr. Yamanaka’s Nobel-winning discovery of iPSCs enabled the creation of patient-specific neural lineages, including motor neurons and glia, offering personalized therapeutic possibilities for HSPs.
First Use of iPSC-Derived Motor Neurons in Spastic Paraplegia Models: Dr. Kevin Eggan, Harvard, 2014
Eggan’s lab successfully differentiated iPSCs into corticospinal motor neurons and modeled axonal degeneration in vitro—providing an unprecedented testing ground for regenerative therapies.
Clinical Trials of Umbilical MSCs in Spinal Motor Disorders: Dr. Ji-Woong Park, South Korea, 2020
Park’s team used umbilical MSCs in pediatric spasticity syndromes with improved motor scores, spasticity reduction, and neuroplastic recovery, setting the stage for broader clinical use in spastic paraplegia [9-12].
Our protocol for Cellular Therapy and Stem Cells for Spastic Paraplegia integrates a dual-route delivery method to ensure robust CNS targeting:
This dual-pathway approach enhances neuroregeneration, improves cell homing, and prolongs therapeutic efficacy for patients with spastic paraplegia [9-12].
At DRSCT, we maintain rigorous ethical standards in sourcing and deploying regenerative products:
These ethical strategies reinforce our mission to deliver safe, transparent, and regenerative care for individuals with spastic paraplegia [9-12].
Preventing the neurodegenerative progression of Spastic Paraplegia (SP) requires early, regenerative, and neuromodulatory interventions. Our protocols are designed to intervene before irreversible corticospinal tract damage occurs. We integrate:
By targeting the upstream mechanisms of neuronal degeneration and spasticity, our Cellular Therapy and Stem Cells for Spastic Paraplegia approach offers a paradigm shift in early intervention and disease modification for hereditary and sporadic forms of SP [13-16].
Our neurology and regenerative medicine experts emphasize the importance of early cellular intervention to preserve functional integrity and motor independence. Initiating stem cell therapy during early-stage SP—before severe axonal degeneration—results in:
Timely initiation of Cellular Therapy and Stem Cells for Spastic Paraplegia maximizes therapeutic efficacy, especially in early-onset or slowly progressive variants of hereditary spastic paraplegia (HSP). Our team ensures early diagnostics, neurofunctional mapping, and intervention planning to optimize outcomes [13-16].
Spastic Paraplegia, particularly the hereditary subtypes (e.g., SPG4, SPG11), is characterized by progressive degeneration of long axons in the corticospinal tracts. Our cellular therapy program addresses these mechanisms through multi-pronged regenerative strategies:
Together, these mechanisms establish a foundation for reversing or halting the progression of neuroaxonal loss in SP and improving neuromuscular control [13-16].
Spastic Paraplegia progresses through definable stages of neurodegeneration, beginning with subtle corticospinal tract dysfunction and advancing toward functional immobility. Cellular therapy offers stage-specific opportunities for intervention:
Stage 1: Subclinical Axonopathy
Stage 2: Functional Gait Impairment
Stage 3: Assistive Device Dependence
Stage 4: Wheelchair Dependence
Stage 5: Advanced Neurological Decline
Stage 1: Subclinical Axonopathy
Stage 2: Functional Gait Impairment
Stage 3: Assistive Device Dependence
Stage 4: Wheelchair Dependence
Stage 5: Advanced Neurological Decline
Our SP-specific regenerative medicine program is rooted in scientific innovation and clinical precision:
By pioneering stem cell applications in SP, we aim to restore neural function, delay disease progression, and ultimately improve independence and neurological resilience [13-16].
By leveraging allogeneic Cellular Therapy and Stem Cells for Spastic Paraplegia, we provide high-efficacy, minimally invasive, and rapid-intervention options for patients across the SP spectrum [13-16].
Our allogeneic stem cell therapy for Hereditary Spastic Paraplegia (HSP) utilizes ethically sourced, high-potency cells designed to target neurodegeneration, reduce spasticity, and promote axonal repair. These include:
Umbilical Cord-Derived Mesenchymal Stem Cells (UC-MSCs): Renowned for their robust proliferation and immunomodulatory properties, UC-MSCs have demonstrated efficacy in reducing muscle spasticity and enhancing motor function in HSP patients.
Wharton’s Jelly-Derived MSCs (WJ-MSCs): These cells are rich in neurotrophic factors and exhibit potent anti-inflammatory effects, making them ideal for mitigating neuroinflammation associated with HSP.
Placental-Derived Stem Cells (PLSCs): PLSCs secrete a variety of growth factors that support neural regeneration and may aid in restoring motor function in HSP patients.
Amniotic Fluid Stem Cells (AFSCs): AFSCs possess the ability to differentiate into neural lineages and contribute to the repair of damaged neural tissues, offering potential benefits in HSP therapy.
Neural Progenitor Cells (NPCs): These cells can differentiate into various neural cell types, providing a direct approach to replacing damaged neurons and supporting neural network restoration in HSP.
By leveraging these diverse allogeneic stem cell sources, our regenerative approach aims to maximize therapeutic potential while minimizing immune rejection [17-18].
Our laboratory adheres to the highest safety and scientific standards to ensure effective stem cell-based treatments for Hereditary Spastic Paraplegia (HSP):
Regulatory Compliance and Certification: Fully registered with the Thai FDA for cellular therapy, following GMP and GLP-certified protocols.
State-of-the-Art Quality Control: Utilizing ISO4 and Class 10 cleanroom environments, we maintain rigorous sterility and quality measures.
Scientific Validation and Clinical Trials: Our protocols are backed by extensive preclinical and clinical research, ensuring evidence-based and continuously refined treatments.
Personalized Treatment Protocols: We tailor stem cell type, dosage, and administration route to each patient’s HSP severity for optimal outcomes.
Ethical and Sustainable Sourcing: Stem cells are obtained through non-invasive, ethically approved methods, supporting long-term advancements in regenerative medicine.
Our commitment to innovation and safety positions our regenerative medicine laboratory as a leader in Cellular Therapy and Stem Cells for Spastic Paraplegia [17-18].
Key assessments for determining therapy effectiveness in HSP patients include spasticity grading (Modified Ashworth Scale), gait analysis, neuroimaging for corticospinal tract integrity, and overall motor function tests. Our Cellular Therapy and Stem Cells for HSP have shown:
Significant Reduction in Muscle Spasticity: MSC-based therapy decreases spasticity by modulating neural pathways and reducing neuroinflammation.
Enhanced Neural Regeneration: Stem cells facilitate the repair of damaged neurons and support the regeneration of neural networks, improving motor function.
Suppression of Inflammatory Pathways: Stem cell therapy modulates pro-inflammatory cytokines, reducing inflammation and oxidative stress in neural tissues.
Improved Quality of Life: Patients experience better mobility, reduced symptoms of spasticity, and enhanced daily functioning.
By providing long-term neuroprotective effects, our protocols for Cellular Therapy and Stem Cells for Spastic Paraplegia offer a revolutionary, evidence-based approach to managing this chronic condition [17-18].
Our team of neurologists and regenerative medicine specialists carefully evaluates each international patient with Hereditary Spastic Paraplegia (HSP) to ensure maximum safety and efficacy in our cellular therapy programs. Due to the progressive nature of HSP and its systemic complications, not all patients may qualify for our advanced stem cell treatments.
We may not accept patients with advanced neurodegeneration characterized by severe mobility impairment, significant cognitive decline, or other comorbidities that may compromise treatment efficacy. Similarly, patients with active infections, uncontrolled systemic diseases, or malignancies are not suitable candidates due to excessive risks.
Additionally, individuals with severe coagulopathies, chronic kidney failure requiring dialysis, or active systemic infections must achieve stabilization before consideration for treatment. Patients with ongoing substance abuse, severe malnutrition, or uncontrolled diabetes must undergo pre-treatment optimization to enhance the success of cellular therapy.
By adhering to stringent eligibility criteria, we ensure that only the most suitable candidates receive our specialized Cellular Therapy and Stem Cells for Spastic Paraplegia, optimizing both safety and therapeutic outcomes [17-18].
Our neurology and regenerative medicine team acknowledges that certain advanced Hereditary Spastic Paraplegia (HSP) patients may still benefit from our Cellular Therapy and Stem Cells for Spastic Paraplegia programs, provided they meet specific clinical criteria. Although the primary goal is to enhance neural regeneration and function, exceptions may be made for patients with rapidly progressing neurodegeneration who remain clinically stable for therapy.
Prospective patients seeking consideration under these special circumstances should submit comprehensive medical reports, including but not limited to:
Neuroimaging: MRI or CT scans to assess neural degeneration and spinal cord integrity.
Neurological Assessments: Evaluations of motor function, spasticity levels, and cognitive status.
Blood Biomarkers: Inflammatory markers (IL-6, TNF-alpha), metabolic panels (HbA1c, cholesterol), and kidney function (BUN, creatinine).
Genetic and Autoimmune Screening: Identifying risk factors for concurrent neurological diseases.
Lifestyle Assessment: Verification of a stable lifestyle with no ongoing substance abuse.
These diagnostic assessments allow our specialists to evaluate the risks and benefits of treatment, ensuring only clinically viable candidates are selected for Cellular Therapy and Stem Cells for Spastic Paraplegia. By leveraging regenerative medicine, we aim to slow disease progression and enhance neural function in eligible patients [17-18].
Ensuring patient safety and optimizing therapeutic efficacy are our top priorities for international patients seeking Cellular Therapy and Stem Cells for Hereditary Spastic Paraplegia (HSP). Each prospective patient must undergo a thorough qualification process conducted by our team of neurologists, regenerative medicine specialists, and metabolic disease experts.
This comprehensive evaluation includes an in-depth review of recent diagnostic imaging (within the last three months), including MRI or CT scans. Additionally, critical blood tests such as complete blood count (CBC), inflammatory markers (CRP, IL-6), metabolic panels (HbA1c, cholesterol), and kidney function tests (creatinine, BUN) are required to assess systemic health and inflammatory status [17-18].
Following a thorough medical evaluation, each international patient receives a personalized consultation detailing their regenerative treatment plan. This includes an overview of the stem cell therapy protocol, specifying the type and dosage of stem cells to be administered, estimated treatment duration, procedural details, and cost breakdown (excluding travel and accommodation expenses).
The primary components of our Cellular Therapy and Stem Cells for Spastic Paraplegia involve the administration of mesenchymal stem cells (MSCs) derived from umbilical cord tissue, Wharton’s Jelly, amniotic fluid, or placental sources. These allogeneic stem cells are introduced via targeted intrathecal injections and intravenous (IV) infusions to enhance neural regeneration, reduce inflammation, and improve motor function.
In addition to Cellular Therapy and Stem Cells for HSP, adjunctive regenerative treatments such as platelet-rich plasma (PRP) therapy, extracellular vesicles (exosomes), growth factors, and anti-inflammatory peptide infusions may be incorporated to optimize therapeutic outcomes. Patients will also receive structured follow-up assessments to monitor neurological improvements and adjust treatment protocols accordingly [17-18].
Once international patients pass our rigorous qualification process, they undergo a structured treatment regimen designed by our regenerative medicine specialists and neurology experts. This personalized protocol ensures the highest efficacy in reducing neural inflammation, promoting neural repair, and improving motor function.
The treatment plan includes the administration of 50-150 million mesenchymal stem cells (MSCs) through a combination of:
Intrathecal Injections: Delivered directly into the cerebrospinal fluid via lumbar puncture to promote neural regeneration and reduce spasticity.
Intravenous (IV) Infusions: Supporting systemic anti-inflammatory effects, immune modulation, and metabolic stabilization.
Exosome Therapy: Enhancing intercellular communication to improve neural function and tissue repair.
The average duration of stay in Thailand for completing our specialized HSP therapy protocol ranges from 10 to 14 days, allowing sufficient time for stem cell administration, monitoring, and supportive therapies. Additional cutting-edge treatments, including hyperbaric oxygen therapy (HBOT), neuro-targeted laser therapy, and metabolic detoxification programs, are integrated to optimize cellular activity and maximize regenerative benefits.
A detailed cost breakdown for our Cellular Therapy and Stem Cells for Spastic Paraplegia ranges from $15,000 to $45,000 [17-18].