INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1350
www.rsisinternational.org
Overcoming Translational Barriers in Neuroprotection: From
Molecular Targets to Biomarkers
Ms. Pooja Suresh Patil
1
, Dr. Md. Javeed Md. Yakub Manure
2
1
Shree Ambabai Talim Sanstha’s Diploma in Pharmacy College, Miraj
2
Appasaheb Birnale College of Pharmacy, Sangli
DOI:
https://doi.org/10.51244/IJRSI.2025.1210000119
Received: 22 October 2025; Accepted: 28 October 2025; Published: 07 November 2025
ABSTRACT
Neuroprotection encompasses strategies aimed at preserving neuronal structure and function by mitigating
pathological processes such as oxidative stress, excitotoxicity, neuroinflammation, and regulated cell death
(RCD). Despite promising preclinical findings, translation to clinical success has been limited due to
pharmacokinetic constraints, inadequate blood-brain barrier (BBB) penetration, methodological deficiencies,
and a lack of objective biomarkers. Key mechanistic targets include mitochondrial stabilization, modulation of
glutamate-induced calcium overload, suppression of proinflammatory glial activation, and inhibition of specific
RCD pathways such as necroptosis (RIPK1/RIPK3/MLKL) and ferroptosis (GPX4/ALOX-15). Advanced in
vitro platformsincluding iPSC-derived organoids and co-culture systemsand rigorously designed in vivo
models are essential for mechanistic validation and prediction of clinical efficacy. Translational success further
relies on achieving therapeutically relevant unbound drug concentrations in the CNS (Cu,br) and integrating
clinically validated biomarkers (e.g., plasma NfL, GFAP) into preclinical endpoints. Emerging therapeutic
strategies encompass CNS-penetrant small molecules, biologics, cell-derived extracellular vesicles, and
phytochemicals delivered via advanced formulations. This review emphasizes the necessity of mechanism-
specific, biomarker-driven, and rigorously validated approaches to overcome historical translational failures and
realize clinically effective neuroprotective interventions.
Keywords: Neuroprotection, Regulated Cell Death, Necroptosis, Ferroptosis, Oxidative Stress,
Neuroinflammation, Blood-Brain Barrier, Translational Biomarkers, iPSC-Derived Organoids, CNS
Therapeutics
Introduction to Neuroprotection and the Translational Imperative
Defining Neuroprotection, Goals, and Scope
Neuroprotection encompasses pharmacological, biological, and nutritional strategies aimed at mitigating or
halting neuronal injury and death. These interventions seek to preserve neuronal integrity and function, thereby
slowing the progression of chronic neurodegenerative diseases such as Alzheimer’s disease (AD) and
Parkinson’s disease (PD), as well as mitigating acute neurological insults including ischemic stroke, traumatic
brain injury (TBI), and spinal cord injury. Despite the heterogeneity of clinical phenotypes, these conditions
share convergent molecular pathologies, notably excitotoxic calcium dysregulation, oxidative stress-induced
mitochondrial dysfunction, neuroinflammatory signaling, and aberrant activation of regulated cell death
pathways, including apoptosis, necroptosis, and ferroptosis. Targeting these mechanisms forms the cornerstone
of contemporary neuroprotective research. The scope of neuroprotective intervention is ambitious. It involves
maintaining cellular homeostasis against these converging pathologies, preventing widespread neuronal loss,
and, ideally, fostering an environment conducive to functional recovery following injury.
(1)
The Crisis of Clinical Translation in Neuroprotection
Despite extensive preclinical success, the clinical translation of neuroprotective agents has been largely
disappointing, constituting a recognized crisis in the field. This failure arises from a complex interplay of
INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1351
www.rsisinternational.org
biological, pharmacological, and methodological limitations.
Preclinical Model Limitations: In vitro systems, including co-cultures and iPSC-derived neuronal models,
often oversimplify human neuropathology and fail to recapitulate the metabolic, vascular, and immune
interactions of the neurovascular unit (NVU)
Pharmacokinetic Constraints: The blood-brain barrier (BBB) presents a significant obstacle, limiting the
delivery of therapeutic agents to the central nervous system (CNS). Compounds demonstrating efficacy in cell
cultures frequently fail to achieve therapeutic concentrations in vivo due to poor BBB penetration
Methodological Deficiencies: Preclinical studies often lack rigorous design elements such as randomization,
blinding, and adherence to standardized reporting guidelines (e.g., STAIR, ARRIVE), leading to inflated
efficacy signals and poor reproducibility
Clinical Trial Challenges: Human studies face additional obstacles, including patient heterogeneity, narrow
therapeutic windows, and the absence of rapid, objective biomarkers for target engagement or neuroprotection
Addressing these systemic barriers is essential to enhance the predictive power of preclinical research and
improve the translational pipeline for neuroprotective therapeutics.
(1)
Core Mechanistic Targets and Regulated Cell Death Pathways
Neuronal viability hinges on tightly regulated energy and immune processes. Neuroprotective strategies are
defined by their ability to interrupt the destructive loops initiated by metabolic and immunological insults.
Oxidative Stress and Mitochondrial Dysfunction
Neurons are highly metabolically active and exceptionally sensitive to oxidative stress, where reactive oxygen
species (ROS) and reactive nitrogen species (RNS) exceed intrinsic antioxidant defenses. Mitochondria are
central to this pathology, functioning both as the primary ROS source (via the electron transport chain) and as a
principal target of oxidative damage. Mitochondrial dysfunction impairs ATP production and triggers
mitochondrial permeability transition pore (mPTP) opening, leading to cytochrome c release and activation of
intrinsic apoptotic cascades. Dysregulation of the Nrf2-ARE pathway further compromises antioxidant defenses,
reducing expression of glutathione, superoxide dismutase, and catalase.
Neuroprotective strategies now focus on enhancing endogenous defenses rather than indiscriminate ROS
scavenging. Nrf2 activators (e.g., dimethyl fumarate, sulforaphane) upregulate antioxidant and detoxification
pathways, while mitochondrial stabilizers (e.g., coenzyme Q10, creatine) preserve mitochondrial integrity and
prevent mPTP opening.
(2)
Neuroprotective efforts targeting oxidative stress have shifted from nonspecific ROS scavengers (like Vitamin
E) to agents that modulate endogenous capacity. Nrf2 activators, such as dimethyl fumarate or sulforaphane,
enhance cellular detoxification and long-term antioxidant defense. Mitochondrial stabilizers, exemplified by
coenzyme Q10 or creatine, aim to preserve function and prevent mPTP opening.
(2)
Excitotoxicity and Calcium Dysregulation
Excitotoxicity results from sustained glutamate receptor (NMDA/AMPA) activation, causing excessive Ca²⁺
influx. Elevated intracellular Ca²⁺ activates destructive enzymes (calpains, caspases) and nitric oxide synthase
(nNOS), exacerbating oxidative stress and mitochondrial depolarization. NMDA receptor antagonists, such as
memantine, show modest clinical benefit but are limited by narrow therapeutic windows and potential disruption
of physiological signaling, highlighting the need for selective modulators of pathological glutamate activity.
(2)
Neuroinflammation, Glial Activation, and Immune Signaling
Neuroinflammation is a complex, double-edged immune response mediated primarily by resident microglia and
INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1352
www.rsisinternational.org
astrocytes. Initially, these cells are protective, engaging in debris clearance and releasing neurotrophic factors
(e.g., BDNF). However, chronic or acute overactivation drives a pathogenic response, characterized by
microglial polarization toward the proinflammatory M1 phenotype, leading to the release of neurotoxic cytokines
(IL-, TNF-α, IL-6) and ROS/RNS, which exacerbates neuronal loss. Reactive astrocytes also contribute by
releasing inflammatory mediators and glutamate.
(2)
A key link between metabolic and inflammatory injury involves the concept of Damage-Associated Molecular
Patterns (DAMPs). Dysfunctional mitochondria produce high levels of ROS, which act as DAMPs, potentially
activating the NLRP3 inflammasome. This activation initiates a strong inflammatory response. Therefore,
successful neuroprotection often requires strategies that promote the anti-inflammatory M2 phenotype in
microglia, suppress the release of proinflammatory cytokines, or directly block DAMP signaling. Agents like
minocycline and natural compounds such as curcumin and resveratrol have demonstrated preclinical efficacy in
reducing glial activation.
(3)
Advanced Regulated Cell Death (RCD) Pathways: Necroptosis and Ferroptosis
The current paradigm in neuroprotection recognizes that cellular demise is often executed through genetically
controlled, non-apoptotic mechanisms, making these pathways attractive, specific therapeutic targets.
(3)
Necroptosis: RIPK1/RIPK3 and the Clinical Pipeline
Necroptosis is a regulated form of necrosis, distinguished by the activation of a signaling complex known as the
necrosome (RIPK1RIPK3MLKL). When caspase-8 activity is inhibited (a common event in severe
ischemia), RIPK1 is phosphorylated and initiates this cascade, ultimately leading to MLKL phosphorylation,
oligomerization, membrane rupture, and uncontrolled release of inflammatory contents (DAMPs). Necroptosis
is implicated in the pathogenesis of TBI, ischemic brain injury, AD, and PD.
(3)
The development of potent, CNS-penetrant RIPK1 inhibitors represents a major translational victory. Small
molecules targeting the allosteric pocket of RIPK1, such as the necrostatins (Nec-1, a tool compound),
demonstrated proof-of-concept in preclinical models of ischemia and neuroinflammation. Building upon this,
Denali Therapeutics and Sanofi developed clinical candidates: SAR443060 (formerly DNL747) is a selective,
orally bioavailable RIPK1 inhibitor that was evaluated in Phase I/Ib studies in healthy subjects and patients with
AD and ALS. Pharmacokinetic-pharmacodynamic (PK-PD) modeling suggested that SAR443060 doses
achieved greater than 80% inhibition of
pRIPK1 (phosphorylated RIPK1) throughout the twice-daily dosing interval, confirming robust target
engagement and CNS penetration. Furthermore, SAR443820 (DNL788), a CNS-penetrant RIPK1 inhibitor,
progressed to a Phase 2 clinical trial for Multiple Sclerosis, confirming the therapeutic potential of blocking this
specific RCD pathway across multiple inflammatory and neurodegenerative conditions. The clinical
advancement of these highly specific RCD inhibitors is a strong indicator that targeting executioner mechanisms
is more viable than utilizing generalized strategies.
(4)
Ferroptosis: The Iron-Dependent Lipid Crisis
Ferroptosis is a recently defined RCD pathway that is dependent on iron and characterized by the catastrophic
accumulation of lipid peroxides. This process is driven by the depletion of the master antioxidant glutathione
(GSH), which subsequently inactivates the lipid repair enzyme glutathione peroxidase 4 (GPX4). The presence
of labile iron, often accumulating in pathological conditions such as TBI, PD, and AD , catalyzes the formation
of highly toxic lipid ROS via Fenton chemistry.
Key molecular markers reflect this pathology. Reduced levels of GSH and GPX4, along with increased
accumulation of the toxic lipid aldehyde 4-hydroxynonenal (4-HNE), are consistently observed in the vulnerable
neuronal populations of PD (substantia nigra, SN) and AD (hippocampus and cortex). Furthermore, elevated
expression of the Transferrin Receptor (TFRC/TfR1) enhances iron influx, and increased levels of Acyl-CoA
INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1353
www.rsisinternational.org
Synthetase Long-Chain Family Member 4 (ACSL4) promote the incorporation of susceptible fatty acids into
membranes, increasing vulnerability to ferroptosis.
(4)
Therapeutic interventions are focused on three strategies: 1) Iron Chelation (e.g., deferoxamine), 2) GPX4
restoration (e.g., N-acetylcysteine), and 3) Inhibition of Lipid Peroxidation (e.g., Ferrostatin-1, Liproxstatin-
1). An important target is the ALOX-15 (15-lipoxygenase) enzyme, which is critical in generating lipid
hydroperoxides. The ALOX-15 specific inhibitor ML351 successfully inhibited cardiomyocyte ferroptosis,
protected injured myocardium, and restored cardiac function following ischemia/reperfusion injury. Similarly,
the anti-ferroptotic reductive lipoxygenase inhibitor PTC-041 demonstrated potent protection against lipid
peroxidation, prevented neuronal loss in primary rat neuronal cultures, and mitigated motor deficits and α-
synuclein aggregation in preclinical PD models (6-OHDA rats and transgenic Line 61 mice). The success of
these specific enzyme inhibitors suggests that targeting the core mechanism of regulated cell deathbe it
necroptosis or ferroptosisyields a more robust and translatable neuroprotective effect than generalized ROS
scavenging.
(5)
Table 1: Key Markers and Inhibitors of Emerging Regulated Cell Death (RCD) in Neurodegeneration
RCD
Pathway
Pathological Mechanism
Disease Links
Preclinical/Clinical Inhibitor
Examples
Ferroptosis
Iron-dependent lipid peroxidation;
GPX4 inactivation; accumulation of
lipid ROS (4-HNE, lipid peroxides)
AD, PD (SN loss),
Ischemic Stroke,
TBI
Ferrostatin-1 (Tool), Iron Chelators
(Deferoxamine), ALOX-15
inhibitors (ML351, PTC-041)
Necroptosis
RIPK1/RIPK3/MLKL activation;
Membrane rupture; DAMP release
ALS, MS, TBI,
Ischemic Stroke,
AD
Necrostatin-1 (Tool), CNS-penetrant
RIPK1 inhibitors
(SAR443060/DNL747,
SAR443820/DNL788)
Apoptosis
Caspase activation; Cytochrome c
release; DNA fragmentation
AD, PD, Stroke
(Penumbra)
Caspase inhibitors, Bcl-2
modulators, Mitochondrial stabilizers
Preclinical Models: Design, Assessment, and Rigor in the CNS
The successful translation of neuroprotective therapies depends on utilizing preclinical models that accurately
reflect human disease pathology and assessing efficacy using rigorous, multimodal methodologies.
In Vitro Screening Platforms: Strengths, Limitations, and Advanced Systems
Traditional Cell Lines vs. Primary Cultures
Neuroprotective compound screening traditionally begins with high-throughput screening (HTS) in
immortalized cell lines due to their ease of culture and scalability. SH-SY5Y cells, derived from human
neuroblastoma, are commonly differentiated for use in PD models, studying toxins like rotenone or MPP+. The
HT22 mouse hippocampal line is particularly valuable for studying oxidative stress independent of
excitotoxicity, as it naturally lacks NMDA receptors, making it an excellent platform for testing ferroptosis
inhibitors or Nrf2 activators under glutathione depletion paradigms.
For studies requiring higher physiological relevance, primary neuronal cultures derived from rodents (e.g.,
cortex, hippocampus, or substantia nigra) are necessary. These systems retain native synaptic function and
morphology, making them ideal for electrophysiology and modeling complex neurotransmitter responses. Co-
culturing primary neurons with primary astrocytes or microglia is crucial for modeling non-cell-autonomous
toxicity and neuroimmune interactions, which are critical in understanding neurodegenerative processes.
(5)
Advanced 3D Modeling: iPSC-Derived Organoids for HTS
A key evolution in neuroprotection research involves the use of patient-derived iPSCs, which can be
INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1354
www.rsisinternational.org
differentiated into specific neuronal subtypes or complex brain organoids (COs). These platforms enable the
modeling of patient-specific genetics (e.g., LRRK2 mutations in PD, APOE variants in AD). This represents a
crucial step toward personalized medicine, as researchers can now screen compounds against a patient’s specific
disease mechanisms.
However, adapting these systems for HTS requires significant methodological optimization. Traditional
organoid generation protocols often involve embedding cells in Matrigel droplets, leading to variability in size,
maturity, and inconsistent focal planes for imaging. To overcome this, researchers are establishing miniaturized
3D culture platforms compatible with 96- or 384-well plates, often using coated microwell plates instead of
Matrigel droplets to allow for automated focus scanning and high-content imaging (HCS). By standardizing size
and maturation protocols, these human-relevant models can transition from being specialized research tools to
efficient HTS platforms, enabling the identification of therapeutic agents tailored to specific genetic
backgrounds, thereby significantly improving the probability of clinical success before costly animal trials are
initiated.
(6)
Essential Multimodal Endpoints for In Vitro Efficacy
Relying on a single endpoint, such as a basic metabolic assay (e.g., MTT), is insufficient and risks yielding
spurious results. Robust in vitro neuroprotection requires a multiparametric approach combining orthogonal
assays:
Viability and Cytotoxicity: LDH release, Resazurin reduction, and Trypan Blue exclusion provide basic
survival data.
Mechanistic Assays: Ca2+ imaging (Fluo-4), mitochondrial membrane potential (ΔΨm via JC-1), and
ATP content define cellular health.
RCD Specificity: Specific markers like Caspase-3/7 activity (for apoptosis) or the GSH/GSSG ratio and
DCFDA (for oxidative RCD/ferroptosis) are necessary to confirm the mechanism of protection.
Functional Assays: Quantification of neurite outgrowth (using HCS) and assessment of network activity
via Multi-Electrode Arrays (MEAs) provide functional metrics that are critical for assessing true
neurorestoration potential.
(6)
In Vivo Modeling: Standardized Paradigms and Functional Readouts
In vivo animal models are essential for integrating pharmacological agents into an intact biological system,
allowing for the evaluation of BBB penetration, metabolism, systemic inflammation, and, critically, functional
recovery.
Acute Ischemic Stroke (MCAO/tMCAO): Rigor and Standardization
The Middle Cerebral Artery Occlusion (MCAO) model in rats or mice is the gold standard for modeling focal
ischemic stroke, particularly the transient variant (tMCAO) which models ischemia-reperfusion injury. The
reliability of this model, however, is exquisitely sensitive to experimental rigor.
The failure of many anti-stroke agents in clinical trials has been directly linked to methodological deficiencies
in preclinical MCAO studies. Guidelines from the Stroke Therapy Academic Industry Roundtable (STAIR) and
the ARRIVE initiative mandate strict control and comprehensive reporting. Key requirements include:
Physiological Monitoring: Maintaining the animal's core temperature (37.0 ± 0.5 °C) and monitoring
critical physiological parameters (blood pressure, blood gases) are vital. A retrospective analysis
demonstrated that, despite the wide adoption of these guidelines, the frequency of reporting temperature,
blood pressure, and blood gas monitoring actually decreased between 2009 and 2019. This continued
deficit in reporting essential procedural quality undermines the reproducibility of results.
Ischemia Verification: The gold standard for confirming successful occlusion and subsequent
reperfusion is Cerebral Blood Flow (CBF) measurement, typically using laser Doppler.
INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1355
www.rsisinternational.org
Surgical Consistency: The choice of surgical tool impacts outcomes; silicone-coated monofilaments are
generally preferred over flame/heat-blunted filaments because they result in lower rates of subarachnoid
hemorrhage (SAH) and less variable infarct volumes.
Unbiased Endpoint Assessment: Infarct volume, typically assessed 24 to 72 hours post-ischemia using
2,3,5-triphenyltetrazolium chloride (TTC) staining, must be calculated with correction for tissue edema.
Furthermore, the practice of a priori sample size calculation, essential for ensuring statistical power, is reported
in less than 10% of studies, suggesting that most preclinical neuroprotection studies are inadequately powered,
further contributing to unreliable findings and the translational pipeline failure.
Table 2: Critical Quality Control Parameters for Translational MCAO Neuroprotection Studies
Experimental
Parameter
Relevance to Reproducibility
Status in Published
Literature (2009-2019)
Core
Temperature
Control
Minimizes variability in infarct
volume; prevents confounding
thermal effects
Reporting decreased
over time
Ischemia
Verification
Gold standard for confirming required
degree of ischemia
Rarely applied
Bias
Minimization
Essential for unbiased data
interpretation (ARRIVE Essential 10)
Increased over time, but
still insufficient
Sample Size
Reporting
Ensures statistical power and validity
Rarely reported (<10%)
Chronic Neurodegeneration and TBI Models
For chronic neurodegenerative disorders, model choice is dictated by the specific pathology under investigation.
Parkinson’s Disease: Toxin models, such as unilateral injection of 6-hydroxydopamine (6-OHDA) or
systemic MPTP administration in mice, induce acute loss of dopaminergic (DA) neurons in the substantia
nigra pars compacta (SNpc). Efficacy must be quantified using immunohistochemistry for tyrosine
hydroxylase (TH+ neuron counts), ideally via stereology, combined with quantitative motor behavioral
readouts (e.g., amphetamine-induced rotations, pole test).
Alzheimer’s Disease: Transgenic models (e.g., APP/PS1, 3xTg-AD, 5xFAD) are used to study amyloid-
beta () plaque burden, tau phosphorylation, and cognitive decline. Key behavioral assays include the
Morris Water Maze (MWM) for spatial learning and memory and the Novel Object Recognition (NOR)
test for recognition memory.
TBI: Models like Controlled Cortical Impact (CCI) or Fluid Percussion Injury (FPI) are used to simulate
blunt trauma, producing focal contusion, edema, and inflammation. Outcomes rely on lesion volumetry
and behavioral assessments (e.g., rotarod, MWM for long-term cognitive deficits).
Advanced Preclinical and Translational Best Practices
To bridge the chasm between preclinical efficacy and clinical success, neuroprotection research must adopt a set
of advanced pharmacological and methodological best practices.
Pharmacokinetics and CNS Exposure: Overcoming the BBB
The Free Drug Hypothesis: Cu,br as the Efficacy Standard
A significant roadblock in drug translation is the historic reliance on non-predictive pharmacokinetic parameters.
INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1356
www.rsisinternational.org
Historically, CNS drug discovery focused on maximizing the total brain/plasma ratio (Kp). However, Kp reflects
both unbound and tissue-bound drug concentration, with the latter being pharmacologically inert.
The Free Drug Hypothesis posits that pharmacological effect is governed by the concentration of the unbound
drug available at the target site within the brain interstitial fluid (Cu,br). Consequently, total brain levels (Kp) or
even BBB permeability measures do not reliably correlate with pharmacodynamic (PD) endpoints or clinical
efficacy. This necessitates measuring the unbound fraction in the brain (fu,br) and calculating Cu,br. While in
vivo microdialysis is the gold standard for Cu,br measurement, less resource-intensive in vitro methodologies
such as equilibrium dialysis of brain homogenates or brain slice incubationscan be used to estimate fu,br,
which is then combined with total brain concentration to yield Cu,br. Importantly, recent advances in specialized
in vitro BBB co-culture models, utilizing endothelial and glial cells, have shown success in accurately predicting
in vivo Cu,br/Cu,pl ratios for a large panel of drugs, enabling earlier selection of candidates with desirable CNS
exposure profiles.
Efflux Transporters and Advanced Delivery Strategies
The BBB acts as a physical and functional barrier. Functionally, it employs efflux transporters, notably P-
glycoprotein (P-gp), which actively extrude many lipophilic drug candidates from the brain back into the
bloodstream. Successful neuroprotective agents must either be poor substrates for these transporters or achieve
plasma concentrations high enough to overcome the efflux mechanism.
For large molecules or those inherently restricted by the BBB, advanced delivery strategies are required :
Nanotechnology: Utilizing carrier materials such as nanoparticles (NPs) or dendrimers allows for
modified transport. Nanoparticles can be surface-functionalized with ligands (peptides, antibodies) or
polyethylene glycol (PEG) to enhance circulation time and facilitate active transport via receptor-
mediated transcytosis (RMT) or adsorptive-mediated transcytosis (AMT). For example, N-acetylcysteine
(NAC) loaded polyamidoamine dendrimers have been developed to suppress neuroinflammation in
cerebral palsy models.
BBB Disruption: While invasive, focused ultrasound or osmotic opening can temporarily increase
permeability to facilitate drug delivery.
Translational Biomarkers: Bridging Preclinical Efficacy to Human Outcomes
A critical factor contributing to translational failure is the use of non-objective, non-quantifiable, and highly
variable preclinical endpoints (e.g., motor scores) that have poor correlation with human clinical outcomes. The
strategic integration of objective fluid biomarkers, quantifiable in both animal models and human patients, is
essential to validate target engagement and disease modification.
Neuronal and Glial Injury Markers (NfL, GFAP, UCH-L1)
These structural protein markers, measured in cerebrospinal fluid (CSF) or peripheral plasma via highly sensitive
assays (e.g., Simoa), provide an objective measure of neuronal and glial damage.
Neurofilament Light (NfL): NfL is a cytoskeleton component released upon axonal injury. Preclinical
models of acute TBI and stroke show robust changes in NfL levels. Clinically, plasma NfL levels
correlate directly with infarct volume, functional outcome, and mortality in stroke patients. Implementing
plasma NfL as a co-primary endpoint in preclinical MCAO studies, correlating drug-induced reductions
in NfL with corresponding reductions in lesion volume, offers a highly objective validation that the
intervention is affecting a human-relevant biological process.
Glial Fibrillary Acidic Protein (GFAP): GFAP is an intermediate filament protein specific to
astrocytes. Elevated plasma GFAP reliably indicates astroglial activation or injury. GFAP, often paired
with UCH-L1 (ubiquitin C-terminal hydrolase L1, a neuronal trauma marker), is used clinically to
INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1357
www.rsisinternational.org
diagnose TBI, correlating with pathoanatomical lesions. GFAP is also an emerging marker for chronic
neuroinflammation in AD.
LPC Lipids: Lysophosphatidylcholines (LPC) are conditional danger-associated molecular patterns
(DAMPs) that induce sterile inflammation. Elevated brain LPC levels are associated with lesion
development and poorer functional outcomes in preclinical ischemic stroke models (tMCAO). While the
correlation between brain and plasma levels requires thorough investigation, plasma LPC shows promise
as a prognostic marker for stroke severity.
Pathological and Synaptic Biomarkers
For chronic neurodegeneration, the use of pathology-specific biomarkers is well established.
Alzheimer’s Disease: Low CSF Aβ42 levels consistently correlate with high fibrillar amyloid plaque
load observed in vivo via PET imaging. Furthermore, plasma and CSF levels of phosphorylated Tau
(pTau) isoforms are now widely accepted as reliable indicators of disease pathology and progression.
Neuroinflammation: CSF levels of soluble TREM2 (sTREM2) are increased in the preclinical stage of
AD, reflecting microglial activation. Utilizing panels of multiplexed cytokines and chemokines (e.g.,IL-
, TNF-α) in CSF allows for detailed monitoring of neuroinflammatory response in trials.
Table 3: Fluid-Based Translational Biomarkers in Acute and Chronic CNS Injury
Biomarker
Cellular Origin /
Pathology
Acute Injury (Stroke/TBI)
Relevance
Chronic Disease (AD/PD)
Relevance
Neurofilament
Light (NfL)
Axonal damage,
Neuronal loss
Strong correlation with
infarct volume and
functional outcome in
patients
Monitors neurodegeneration
rate in PD, AD, MS
Glial Fibrillary
Acidic Protein
(GFAP)
Astrocyte activation /
Injury
TBI diagnostic; reflects
acute glial reactivity
Early stage AD marker;
reflects astroglial pathology
UCH-L1
Neuronal cytoplasm /
Axonal injury
Early marker of acute
neuronal trauma (TBI
diagnostic panel)
Potential relevance to PD,
though plasma correlation is
inconclusive
Phospho-Tau
(pTau)
Neurofibrillary
tangles; synaptic
dysfunction
Minimal acute use
Highly specific for AD
pathology; correlates with PET
amyloid burden
Representative Therapeutic Classes and Clinical Trajectories
Small Molecules and Repurposed Agents
Small molecules remain the backbone of pharmacological neuroprotection due to their advantageous synthesis,
formulation, and dosing control. The clinical failure history, however, provides vital lessons regarding the
necessity of rigor and mechanistic specificity. Translational Failure Analysis: NXY-059 and Nerinetide (NA-1)
The failures of large-scale stroke trials, such as the SAINT II trial for the radical scavenger NXY-059, underscore
the cost of compromised methodology. Analysis showed that the robust efficacy seen in preclinical studies could
not be reproduced across multiple independent academic institutions and was confounded by publication bias
and a failure to rigorously test clinically relevant time windows. More recently, the neuroprotective agent
Nerinetide (NA-1), which targets PSD-95 protein interactions, failed to meet its primary functional outcome
endpoint (mRS 02 at 90 days) in the overall cohort of acute ischemic stroke patients undergoing endovascular
therapy (ESCAPE-NA1 trial). However, a critical subgroup analysis revealed a statistically significant benefit
in functional outcomes, mortality, and infarct size among patients who
INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1358
www.rsisinternational.org
did not receive the standard-of-care thrombolytic agent alteplase. This finding is highly significant: it suggests
that neuroprotection is biologically achievable in humans but that efficacy is critically dependent on patient
stratification and may be negated by adverse drug-drug interactions (DDI) with concurrent therapies. Future
preclinical validation studies must therefore explicitly test the candidate neuroprotective agent in combination
with current clinical standards of care to identify potential inhibitory interactions, thereby preventing conditional
efficacy failures in later clinical phases.
Biologics and Cell-Based Therapies
Biologics offer potent, mechanism-specific interventions but face significant hurdles related to systemic delivery
and immunogenicity.
Neurotrophic Factor Delivery (GDNF Gene Therapy)
Glial Cell Line-Derived Neurotrophic Factor (GDNF) is highly protective for DA neurons but cannot cross the
BBB effectively. To overcome this, strategies involving gene therapy are being pursued. A viral vector, AAV2-
GDNF, is designed to encode the trophic factor and is delivered directly to the putamen of PD patients via
bilateral stereotactic convection-enhanced delivery. This delivery method aims for sustained, localized
expression of GDNF. This approach has advanced to Phase 2 trials (REGENERATE-PD), demonstrating a
favorable safety profile and continued positive trends in clinical measures for moderate-stage PD.
Stem CellDerived Extracellular Vesicles (EVs)
Extracellular vesicles (EVs), including exosomes, are naturally occurring nanocarriers that transfer molecular
cargo (proteins, mRNA, microRNAs) between cells. EVs derived from stem cells, such as neural stem cells
(NSC EVs), are being explored as drug delivery systems and as therapies themselves, carrying neuroprotective
factors (e.g., miRNA-30d/30a) that can modulate neuroinflammation and cell signaling in recipient cells.
Preclinical studies demonstrated that acute intravenous administration of NSC EVs in a rat TBI model resulted
in neuroprotection, evidenced by reduced lesion volume and enhanced motor function recovery. A crucial
finding in these studies was the observation of a gender-dependent therapeutic effect, with significant benefits
in motor function primarily noted in male rats. This highlights the necessity of addressing sex as a biological
variable in preclinical neuroprotection studies, analyzing outcomes in both sexes to avoid misleading efficacy
conclusions that are relevant to only a single population subgroup.
Natural Products and Phytochemicals
Natural products, such as flavonoids like Quercetin and Resveratrol, are attractive due to their pleiotropic
mechanisms, including potent antioxidant and anti-inflammatory properties. Preclinical evidence supports their
role in mitigating secondary damage, such as cerebral edema following TBI (e.g., Quercetin).
However, their translation is severely hampered by poor pharmacokinetics. Many neuroprotective
phytochemicals exhibit very low BBB permeability in vivo. While compounds like Quercetin show low but
measurable brain penetration, others like Resveratrol and Fisetin exhibit very low permeability, failing to reach
therapeutic Cu,br levels when administered systemically. To leverage the inherent neuroprotective benefits of
these compounds, sophisticated formulation strategies, such as liposomal or nanoparticle encapsulation, must be
employed to facilitate RMT/AMT across the BBB.
CONCLUSION AND FUTURE DIRECTIONS
The journey to developing clinically viable neuroprotective agents is characterized by profound mechanistic
challenges and historical methodological failings. However, the field is undergoing a critical, positive pivot
rooted in enhanced scientific rigor and sophisticated target identification.
The future success of neuroprotection hinges on four actionable recommendations:
INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1359
www.rsisinternational.org
1. Prioritize Regulated Cell Death Inhibition: Therapeutic development must focus on specific,
executioner pathways like Necroptosis (RIPK1/RIPK3) and Ferroptosis (GPX4/ALOX-15). The
advancement of CNS-penetrant RIPK1 inhibitors into Phase 2 trials demonstrates that specific blockade
of RCD pathways offers a robust strategy that overcomes the limitations of generalized antioxidant
approaches.
2. Mandate Rigorous Methodological Standards: The pervasive deficits in methodological reporting in
preclinical literature must be resolved. Full adherence to STAIR and ARRIVE guidelines, including
mandatory reporting of physiological parameters (temperature, CBF, BP) and performing a priori sample
size calculations, is essential to generate reproducible and translationally meaningful data, limiting the
high variability that obscured efficacy in historic trials.
3. Optimize Pharmacokinetics for Cu,br: Candidate selection must move beyond the non-predictive Kp
ratio. Compounds must be designed or delivered (via RMT, NPs, or gene therapy) to achieve quantifiable,
therapeutic unbound concentrations (Cu,br) in the brain interstitial fluid, ensuring the drug reaches the
molecular target at an effective level.
4. Integrate Translational Biomarkers into Preclinical Endpoints: Preclinical studies must adopt
clinically validated fluid biomarkers, such as plasma NfL and GFAP, as critical quantitative outcome
measures. Correlating reductions in these circulating markers of neuronal/glial injury with lesion size
and functional recovery provides an objective, clinically relevant validation of the neuroprotective effect,
significantly de-risking the subsequent clinical trial phase.
By adopting this disciplined, biomarker-driven, and mechanism-specific approach, the neurotherapeutics
community can overcome the translational hurdles and finally realize the promise of true neuroprotection.
REFERENCES
1. Verma M, et al. Excitotoxicity, calcium and mitochondria: a triad in synaptic dysfunction. Front Aging
Neurosci. 2022;14:878812. doi:10.3389/fnagi.2022.878812.
2. Dash UC, et al. Oxidative stress and inflammation in the pathogenesis of neurodegenerative diseases.
Science Progress. 2025;108(1):003685042211074. doi:10.1177/003685042211074.
3. Wu W, et al. Molecular mechanisms of excitotoxicity and their relevance to neurodegenerative
diseases. Signal Transduct Target Ther. 2025;10(1):114. doi:10.1038/s41401-025-01576-w.
4. Cui J, et al. Regulated cell death: discovery, features and implications for neurodegenerative diseases.
Biosignaling Funct Genomics. 2021;22(2):115. doi:10.1186/s12964-021-00799-8.
5. Jurcău MC, et al. The link between oxidative stress, mitochondrial dysfunction, and
neurodegeneration. Antioxidants (Basel). 2022;11(11):2167. doi:10.3390/antiox11112167.
6. Dirnagl U, Endres M. Translational stroke research: vision and reality. Stroke. 2014;45(4):12961302.
doi:10.1161/STROKEAHA.114.003959.
7. Macleod MR, et al. Good laboratory practice: preventing introduction of bias at the bench. Stroke.
2008;39(10):28252830. doi:10.1161/STROKEAHA.108.516044.
8. Savitz SI, et al. Translational strategies for neuroprotection in acute ischemic stroke. Nat Rev Neurol.
2017;13:258270. doi:10.1038/nrneurol.2017.37.
9. Ghosh S, Basu A. Preclinical models of neurodegeneration: challenges and opportunities. Front
Neurosci. 2021;15:654951. doi:10.3389/fnins.2021.654951.
10. O’Collins VE, et al. 1,026 experimental treatments in acute stroke. Ann Neurol. 2006;59(3):467477.
doi:10.1002/ana.20741.
11. Fisher M, Saver JL. Future directions of neuroprotection in acute ischemic stroke. Stroke.
2015;46(4):11671173. doi:10.1161/STROKEAHA.114.007004.
12. Wen P, et al. Oxidative stress and mitochondrial impairment: key drivers of neurodegeneration.
Neurochem Int. 2025. Available from: https://www.sciencedirect.com/.
13. Biswas K, et al. A review on the cell signaling pathways involved in microglial activation and
neuroinflammation. J Neuroinflammation. 2023. Available from: https://pubmed.ncbi.nlm.nih.gov/.
INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1360
www.rsisinternational.org
14. Fei Y, et al. The role of ferroptosis in neurodegenerative diseases. Front Neurosci. 2024; Available
from: https://www.frontiersin.org/.
15. Catalani E, et al. Targeting mitochondrial dysfunction and oxidative stress in retinal pathologies linked
to retinal ganglion cell degeneration. Front Mol Biosci. 2023; Available from:
https://www.frontiersin.org/.
16. Khan S, et al. Excitotoxicity, oxytosis/ferroptosis, and neurodegeneration. Front Neurosci. 2024;
Available from: https://www.frontiersin.org/.
17. Gao C, et al. Microglia in neurodegenerative diseases: mechanism and therapeutic implications. Front
Aging Neurosci. 2023; Available from: https://www.frontiersin.org/.
18. Zong Y, et al. Mitochondrial dysfunction: mechanisms and advances in therapeutic strategies. Nat
Rev Neurol. 2024; Available from: https://www.nature.com/.
19. Ye K, et al. The double-edged functions of necroptosis in neurodegeneration. Cell Death Dis. 2023;
Available from: https://www.nature.com/.
20. Zheng X, et al. Ferroptosis: a novel regulated cell death participating in neurodegeneration. Environ
Health Prev Med. 2023; Available from: https://ehoonline.biomedcentral.com/.
21. Nam D, et al. Effects of calcium ion dyshomeostasis and neuroinflammation in neurodegenerative
diseases. Front Neurosci. 2024; Available from: https://www.frontiersin.org/.
22. Webber EK, et al. Cytosolic calcium: judge, jury, and executioner of neurodegeneration. Alzheimers
Dement. 2023; Available from: https://alz-journals.onlinelibrary.wiley.com/.
23. Xu Y, et al. New insight on microglia activation in neurodegenerative diseases. Front Neurosci. 2023;
Available from: https://www.frontiersin.org/.
24. Liu Y, et al. The research landscape of ferroptosis in neurodegenerative diseases. Front Neurosci.
2024; Available from: https://www.frontiersin.org/.
25. Friedrich B, Lindauer U. Procedural and methodological quality in preclinical stroke research a
cohort analysis of the rat MCAO model comparing periods before and after the publication of
STAIR/ARRIVE. Front Neurol. 2022;13. Available from:
https://pmc.ncbi.nlm.nih.gov/articles/PMC9190283/.
26. Dunkel P, et al. Clinical utility of neuroprotective agents in neurodegenerative diseases: current status
of drug development for Alzheimer’s, Parkinson’s, Huntington’s, and ALS. Expert Opin Investig
Drugs. 2012;21(9):12671308.
27. Di Marco S, et al. Drug metabolism and pharmacokinetics, the blood-brain barrier, and CNS drug
discovery. Front Pharmacol. 2021;12. Available from:
https://pmc.ncbi.nlm.nih.gov/articles/PMC8280005/.
28. O’Collins VE, Macleod MR. A critical appraisal of the NXY-059 neuroprotection studies for acute
stroke: a need for more rigorous testing of neuroprotective agents in animal models. Stroke.
2007;38(4):11211125. doi:10.1161/01.STR.0000251427.00957.17.
29. Percie du Sert N, et al. The ARRIVE guidelines 2.0: updated guidelines for reporting animal research.
PLoS Biol. 2020;18(7):e3000411. doi:10.1371/journal.pbio.3000411.
30. Rauf A, et al. Implications of iron in ferroptosis, necroptosis, and pyroptosis as potential players in
TBI morbidity and mortality. J Inflamm Res. 2023;16:19671977. Available from:
https://pmc.ncbi.nlm.nih.gov/articles/PMC11529200/.
31. Zhuo J, et al. Targeting ferroptosis for neuroprotection: potential therapeutic avenues in
neurodegenerative and neuropsychiatric diseases. Acta Neuropathol Commun. 2024;12(1):41.
Available from: https://pmc.ncbi.nlm.nih.gov/articles/PMC12423104/.
32. Minnella A, et al. Targeting ferroptosis with the lipoxygenase inhibitor PTC-041 as a therapeutic
strategy for Parkinson's disease. PLoS One. 2024;19(4):e0309893. Available from:
https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0309893.
33. Thapa K, et al. Therapeutic insights on ferroptosis in Parkinson's disease. Eur J Pharmacol.
2022;930:175184. Available from:
https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2024.1407335/full.
34. Park S, Mook-Jung I. Brain organoids are new tool for drug screening of neurological diseases. BMB
Rep. 2021;54(8):411420. Available from: https://pmc.ncbi.nlm.nih.gov/articles/PMC8341697/.
35. Durens M, et al. High-throughput screening of human induced pluripotent stem cell-derived brain
organoids. J Vis Exp. 2020;(161). Available from:
INTERNATIONAL JOURNAL OF RESEARCH AND SCIENTIFIC INNOVATION (IJRSI)
ISSN No. 2321-2705 | DOI: 10.51244/IJRSI | Volume XII Issue X October 2025
Page 1361
www.rsisinternational.org
https://www.researchgate.net/publication/339033922_High-
throughput_screening_of_human_induced_pluripotent_stem_cell-derived_brain_organoids.
36. Takhsha A, et al. Neuroprotective potential of phytochemicals. Front Biosci (Elite Ed).
2011;3(4):11421163. Available from: https://pmc.ncbi.nlm.nih.gov/articles/PMC3459459/.
37. Ahangari G, et al. Quercetin protects blood-brain barrier integrity and maintains microvascular
permeability following traumatic brain injury. J Neurosurg Anesthesiol. 2025;37(2):224231.
Available from: https://pubmed.ncbi.nlm.nih.gov/40739078/.
38. Rzeska Z, et al. Much more than nutrients: the protective effects of nutraceuticals on the bloodbrain
barrier in diseases. Nutrients. 2025;17(5):766. Available from: https://www.mdpi.com/1999-
4923/17/5/766.