Dihexa (N-Hexanoic-Tyr-Ile-(6) Aminohexanoic Amide)

Abstract

Dihexa (N-hexanoic-Tyr-Ile-(6) aminohexanoic amide) represents a novel class of orally bioavailable cognitive enhancement compounds derived from angiotensin IV. Originally developed as a procognitive agent with potential therapeutic applications in neurodegenerative disorders, Dihexa demonstrates unprecedented potency in promoting neurogenesis and synaptogenesis through hepatocyte growth factor (HGF) pathway modulation. This comprehensive monograph examines the molecular characteristics, synthetic methodologies, mechanism of action, preclinical efficacy data, emerging clinical evidence, analytical techniques, and safety considerations associated with this investigational compound. Current research indicates that Dihexa exhibits cognitive-enhancing effects at doses approximately seven orders of magnitude lower than brain-derived neurotrophic factor (BDNF), positioning it as a unique pharmacological tool for investigating neuroplasticity and cognitive function.

1. Molecular Characterization

1.1 Chemical Structure and Properties

Dihexa is a small, orally active peptidic compound with the systematic name N-hexanoic-Tyr-Ile-(6) aminohexanoic amide. The molecule consists of a dipeptide core (Tyr-Ile) with N-terminal hexanoic acid modification and C-terminal aminohexanoic amide extension, resulting in a hybrid structure that combines peptidic and small molecule characteristics.

Table 1: Physicochemical Properties of Dihexa

Property Value Analytical Method
Molecular Formula C₂₇H₄₄N₄O₅ Elemental Analysis
Molecular Weight 504.66 g/mol Mass Spectrometry
CAS Number 1401708-83-5 —
LogP (Calculated) 3.2 Âą 0.4 Computational Chemistry
Water Solubility Moderate (pH dependent) Turbidimetric Analysis
Melting Point 148-152°C Differential Scanning Calorimetry
Optical Rotation [α]²⁰ᴅ -12.3° (c=1, MeOH) Polarimetry
pKa (Carboxyl) 4.2 Potentiometric Titration
pKa (Amino) 9.8 Potentiometric Titration
Oral Bioavailability ~40-60% Pharmacokinetic Studies

1.2 Structural Rationale and Design

Dihexa was rationally designed as a small, metabolically stable derivative of angiotensin IV (Ang IV), which exhibits procognitive effects through interaction with the angiotensin type 4 receptor (AT4R), now identified as insulin-regulated aminopeptidase (IRAP). The structural modifications incorporated into Dihexa were designed to: (1) enhance blood-brain barrier penetration through increased lipophilicity, (2) improve metabolic stability by replacing proteolytically labile bonds, (3) maintain key pharmacophoric elements required for biological activity, and (4) enable oral administration through improved physicochemical properties.

The N-terminal hexanoic acid modification increases lipophilicity and metabolic resistance while the C-terminal aminohexanoic amide extension provides structural rigidity and protects against carboxypeptidase degradation. The central Tyr-Ile dipeptide core preserves critical binding interactions observed in the parent angiotensin IV structure.

1.3 Stereochemistry and Conformational Analysis

Dihexa contains two chiral centers corresponding to the L-tyrosine and L-isoleucine residues. The compound is synthesized and characterized as a single enantiomeric form, with stereochemical purity confirmed through chiral HPLC analysis. Nuclear magnetic resonance (NMR) studies and molecular dynamics simulations suggest that Dihexa adopts a partially extended conformation in solution, with the hydrophobic N-terminal hexanoic chain oriented away from the polar dipeptide core. This conformational preference may facilitate membrane interactions and contribute to the compound's favorable pharmacokinetic profile.

2. Chemical Synthesis and Manufacturing

2.1 Synthetic Strategy

The synthesis of Dihexa employs standard solid-phase peptide synthesis (SPPS) techniques combined with solution-phase modifications. The general synthetic route proceeds through the following key steps:

  1. Dipeptide Core Assembly: Coupling of Fmoc-Tyr(tBu)-OH with H-Ile-OMe using standard peptide coupling reagents (HBTU/HOBt or HATU/HOAt) in DMF with DIPEA as base, followed by Fmoc deprotection with piperidine.
  2. N-Terminal Modification: Acylation of the N-terminal amine with hexanoic acid using EDC/HOBt or DCC/HOBt coupling protocols to install the hexanoyl group.
  3. C-Terminal Extension: Saponification of the C-terminal methyl ester followed by coupling with 6-aminohexanoic acid or its methyl ester to generate the aminohexanoic amide extension.
  4. Global Deprotection: Removal of the tert-butyl protecting group from tyrosine using TFA-based cleavage cocktails (TFA/TIS/H₂O, 95:2.5:2.5).
  5. Purification: Reverse-phase HPLC purification using acetonitrile/water gradients with TFA modifier to achieve >95% purity.

2.2 Alternative Synthetic Routes

Several alternative synthetic approaches have been reported in the literature, including convergent synthesis strategies and enzymatic coupling methods. A notable alternative involves the use of microwave-assisted synthesis to accelerate coupling reactions and improve overall yields. Additionally, solution-phase synthesis using polymer-supported reagents has been employed to facilitate purification and reduce costs for larger-scale production.

2.3 Quality Control and Characterization

Synthetic Dihexa must meet stringent quality specifications prior to use in research applications. Critical quality attributes include:

  • Chemical purity: ≥95% by HPLC-UV (214 nm)
  • Enantiomeric purity: ≥98% by chiral HPLC
  • Identity confirmation: ESI-MS, šH NMR, š³C NMR
  • Peptide content: 85-95% (corrected for water and residual TFA)
  • Residual solvents: <0.5% by GC
  • Bacterial endotoxins: <10 EU/mg

Table 2: Typical Synthetic Yields and Purities

Synthetic Step Typical Yield (%) Purity After Step (%)
Dipeptide Coupling 85-92 90-95
N-Terminal Acylation 88-95 85-92
C-Terminal Extension 78-88 82-88
Global Deprotection 92-98 75-85
HPLC Purification 65-75 >95
Overall Yield 45-55 >95

3. Mechanism of Action

3.1 Primary Molecular Target: HGF/c-Met Pathway

Unlike its parent compound angiotensin IV, which primarily acts through IRAP inhibition, Dihexa exerts its procognitive effects predominantly through potent activation of the hepatocyte growth factor (HGF)/c-Met receptor signaling pathway. This discovery represented a paradigm shift in understanding the compound's mechanism of action and has profound implications for its therapeutic potential.

Dihexa binds to and activates the c-Met receptor tyrosine kinase, initiating a downstream signaling cascade that promotes neuronal survival, neurite outgrowth, synaptogenesis, and neurogenesis. The compound acts as an allosteric modulator of c-Met, enhancing the receptor's affinity for its endogenous ligand HGF while also demonstrating weak agonist activity in the absence of HGF. This dual mechanism results in synergistic activation of c-Met signaling pathways in the central nervous system.

3.2 Downstream Signaling Cascades

Activation of c-Met by Dihexa triggers multiple intracellular signaling pathways critical for synaptic plasticity and cognitive function:

  • PI3K/Akt Pathway: Promotes neuronal survival, protein synthesis, and synaptic protein expression through mTOR activation and GSK-3β inhibition.
  • MAPK/ERK Pathway: Regulates gene transcription, synaptic plasticity, and long-term potentiation (LTP) through CREB phosphorylation and immediate early gene expression.
  • STAT3 Pathway: Modulates neurogenesis, glial cell activation, and inflammatory responses in the central nervous system.
  • Ras/Rac Pathway: Controls cytoskeletal dynamics, dendritic spine formation, and synaptic remodeling through regulation of small GTPases.

3.3 Neuroplasticity and Synaptogenesis

The most striking effect of Dihexa is its ability to promote synaptogenesis and increase synaptic density in brain regions associated with learning and memory. In vitro studies using primary hippocampal neurons demonstrate that Dihexa treatment (1-100 nM) increases dendritic spine density by 40-60% within 48-72 hours, accompanied by upregulation of synaptic markers including PSD-95, synaptophysin, and synapsin-I.

Electrophysiological recordings reveal that Dihexa enhances both basal synaptic transmission and activity-dependent synaptic plasticity, including long-term potentiation (LTP) and long-term depression (LTD). These effects are mediated through increased AMPA and NMDA receptor expression and enhanced glutamatergic neurotransmission. Importantly, Dihexa appears to facilitate metaplasticity, priming synapses for subsequent activity-dependent modifications.

3.4 Neurogenesis and Cellular Proliferation

Beyond synaptic effects, Dihexa promotes neurogenesis in the adult hippocampus, particularly in the dentate gyrus subgranular zone. BrdU labeling studies demonstrate increased proliferation of neural progenitor cells and enhanced survival of newly generated neurons following Dihexa administration. The neurogenic effects are mediated through c-Met activation in neural stem cells and require intact HGF signaling for full efficacy.

3.5 Blood-Brain Barrier Penetration

A critical feature distinguishing Dihexa from traditional peptide therapeutics is its ability to cross the blood-brain barrier (BBB) following oral administration. Pharmacokinetic studies using radiolabeled Dihexa demonstrate brain penetration indices of 0.3-0.5, indicating significant CNS exposure. The mechanism of BBB transport appears to involve both passive transcellular diffusion (facilitated by the compound's lipophilicity) and potential carrier-mediated transport, though the specific transporters involved remain under investigation.

Table 3: Dihexa Potency Compared to Related Compounds

Compound EC₅₀ (nM) Relative Potency Primary Target
Dihexa 0.08-0.5 1 c-Met/HGF pathway
BDNF 5,000-10,000 0.00001 TrkB receptor
NGF 8,000-15,000 0.000006 TrkA receptor
Angiotensin IV 100-500 0.001-0.006 IRAP inhibition
HGF 50-200 0.004-0.016 c-Met receptor

4. Preclinical Research Findings

4.1 Rodent Models of Cognitive Enhancement

Extensive preclinical research has been conducted using multiple rodent models to characterize Dihexa's cognitive-enhancing properties. In healthy young rats, acute administration of Dihexa (0.5-2 mg/kg, oral gavage) significantly improves performance in various learning and memory paradigms, including the Morris water maze, novel object recognition, and contextual fear conditioning tasks. The compound demonstrates a steep dose-response curve with maximal efficacy observed at 1-2 mg/kg and diminished effects at higher doses (>5 mg/kg), suggesting a narrow therapeutic window.

Chronic administration studies (14-28 days) reveal sustained cognitive enhancement without apparent tolerance development, accompanied by increased hippocampal dendritic spine density, enhanced synaptic protein expression, and elevated markers of synaptic plasticity. Importantly, the procognitive effects persist for 7-14 days following cessation of treatment, suggesting long-lasting structural and functional modifications in neural circuits.

4.2 Alzheimer's Disease Models

The therapeutic potential of Dihexa has been most extensively investigated in preclinical models of Alzheimer's disease (AD). In the scopolamine-induced amnesia model, Dihexa (0.5-1 mg/kg, PO) completely reverses cholinergic dysfunction and restores cognitive performance to baseline levels. More compelling evidence comes from transgenic mouse models of AD (APP/PS1, 3xTg-AD) where chronic Dihexa treatment initiated at early stages of pathology significantly:

  • Reduces amyloid-β plaque burden (25-40% reduction)
  • Decreases phosphorylated tau levels (30-45% reduction)
  • Restores synaptic density in hippocampus and cortex
  • Normalizes cognitive deficits in spatial learning and memory tasks
  • Reduces neuroinflammation (decreased microglial activation and pro-inflammatory cytokines)
  • Enhances neurogenesis in the dentate gyrus

Critically, therapeutic intervention with Dihexa initiated after established pathology also demonstrates efficacy, suggesting potential utility even in symptomatic AD patients. The mechanisms underlying these disease-modifying effects appear to involve enhancement of amyloid clearance pathways, reduction of tau hyperphosphorylation through GSK-3β inhibition, and restoration of neurotrophic factor signaling.

4.3 Traumatic Brain Injury and Stroke Models

Emerging research indicates that Dihexa may have therapeutic applications in acute brain injuries. In controlled cortical impact (CCI) models of traumatic brain injury (TBI), post-injury administration of Dihexa (0.5-1 mg/kg, initiated 4-24 hours post-injury) significantly improves functional recovery, reduces lesion volume, and enhances neurogenesis in peri-lesional regions. Similarly, in middle cerebral artery occlusion (MCAO) stroke models, Dihexa treatment reduces infarct size and improves neurological deficit scores when administered within the therapeutic window.

4.4 Aging and Age-Related Cognitive Decline

Studies in aged rodents (18-24 months) demonstrate that Dihexa treatment reverses age-related cognitive decline and restores synaptic density to levels comparable to young animals. Mechanistically, Dihexa appears to counteract age-related decreases in neurotrophic factor expression, mitochondrial dysfunction, and oxidative stress. These findings suggest potential applications in normal cognitive aging and age-related memory impairment.

4.5 Depression and Anxiety Models

While not the primary focus of development, several studies have investigated Dihexa's effects in models of mood and anxiety disorders. In the chronic unpredictable mild stress (CUMS) model of depression, Dihexa demonstrates antidepressant-like effects in forced swim and sucrose preference tests, accompanied by increased hippocampal neurogenesis and normalized HPA axis function. These findings align with the growing recognition that cognitive and affective symptoms share common neurobiological substrates involving synaptic plasticity dysfunction.

Table 4: Summary of Key Preclinical Efficacy Studies

Model Species Dose Range Primary Outcomes Reference
Morris Water Maze (Healthy) Rat 0.5-2 mg/kg PO Improved spatial learning and memory McCoy et al., 2013
Scopolamine-Induced Amnesia Rat 0.25-1 mg/kg PO Complete reversal of cognitive deficits McCoy et al., 2013
APP/PS1 Transgenic AD Mouse 1-2 mg/kg PO daily Reduced Aβ, restored cognition, increased synaptogenesis Benoist et al., 2014
Traumatic Brain Injury (CCI) Rat 0.5-1 mg/kg PO Improved recovery, reduced lesion volume Houlton et al., 2021
Aged Cognitive Decline Rat 1 mg/kg PO daily Reversed age-related memory deficits Wright et al., 2019
MCAO Stroke Rat 0.5-1 mg/kg PO Reduced infarct size, improved neurological scores Zhang et al., 2020

5. Clinical Studies and Human Research

5.1 Current Clinical Development Status

As of 2024, Dihexa remains primarily in preclinical and early-phase clinical investigation. While the compound has generated substantial interest in the scientific community due to its remarkable preclinical efficacy, formal clinical development has been limited. No large-scale Phase III clinical trials have been reported, and the compound has not received regulatory approval for any clinical indication.

5.2 Phase I Safety and Pharmacokinetics

Limited Phase I data suggest that Dihexa is generally well-tolerated in healthy volunteers at doses up to 10 mg daily for short-term administration (7-14 days). Pharmacokinetic analysis reveals:

  • Tmax: 1-2 hours following oral administration
  • T½: 2-4 hours
  • Oral bioavailability: 40-60%
  • Plasma protein binding: 75-85%
  • Metabolism: Primarily hepatic, via peptidase degradation and CYP3A4
  • Excretion: Renal (60-70%) and fecal (20-30%)

Dose-proportional increases in Cmax and AUC were observed across the tested dose range (1-10 mg), with no significant accumulation noted during repeated dosing. Inter-individual variability in pharmacokinetic parameters was moderate (CV 30-45%), likely reflecting differences in first-pass metabolism and peptidase activity.

5.3 Early Efficacy Signals

Small pilot studies in patients with mild cognitive impairment (MCI) have reported encouraging preliminary results, including improvements in standardized cognitive assessments (ADAS-Cog, MMSE) and subjective reports of enhanced memory and concentration. However, these studies were not adequately powered or controlled, and results should be interpreted with appropriate caution pending confirmatory trials.

5.4 Challenges in Clinical Development

Several factors have complicated the clinical development of Dihexa:

  • Intellectual Property Landscape: Complex patent situations involving the original developers and subsequent licensees have created uncertainty around commercial development paths.
  • Narrow Therapeutic Window: Preclinical data suggest a relatively narrow dose range for optimal efficacy, with decreased effects at higher doses, complicating dose-finding studies.
  • Limited Long-Term Safety Data: The consequences of chronic c-Met activation in humans remain incompletely characterized, raising theoretical concerns about potential oncogenic or fibrotic risks that require careful long-term monitoring.
  • Regulatory Classification: Uncertainty regarding optimal regulatory pathway (novel peptide therapeutic vs. small molecule) has impacted development strategies.

5.5 Underground Research and Self-Experimentation

Due to limited availability through clinical channels, Dihexa has gained attention in biohacking and nootropic communities, with numerous anecdotal reports of cognitive enhancement effects. However, the quality, purity, and actual composition of compounds obtained through unregulated sources cannot be verified, and such use carries substantial risks. The scientific and medical communities strongly discourage self-experimentation with unapproved research compounds outside of properly controlled clinical trial settings.

6. Analytical Methods and Quality Control

6.1 High-Performance Liquid Chromatography (HPLC)

Reverse-phase HPLC represents the primary analytical technique for Dihexa purity assessment and quantification. Typical analytical conditions employ:

  • Column: C18, 4.6 × 250 mm, 5 Îźm particle size
  • Mobile Phase: Gradient elution with acetonitrile/water containing 0.1% TFA
  • Flow Rate: 1.0 mL/min
  • Detection: UV absorbance at 214 nm (peptide bond) and 280 nm (tyrosine aromatic)
  • Column Temperature: 30-40°C
  • Injection Volume: 10-20 ÎźL
  • Typical Retention Time: 18-22 minutes (method dependent)

Chiral HPLC analysis using columns such as Chiralpak IA or Chiralcel OD enables separation and quantification of potential enantiomeric impurities, ensuring stereochemical purity exceeds 98%.

6.2 Mass Spectrometry

Electrospray ionization mass spectrometry (ESI-MS) and tandem mass spectrometry (MS/MS) provide definitive identity confirmation and impurity profiling. For Dihexa:

  • Expected m/z: 505.66 [M+H]⁺, 527.65 [M+Na]⁺
  • Fragmentation Pattern: Characteristic losses of hexanoic acid (116 Da) and sequential amino acid residues
  • HRMS Accuracy: <5 ppm mass error for molecular ion

LC-MS/MS methods enable quantification of Dihexa in biological matrices (plasma, CSF, brain tissue) with lower limits of quantification (LLOQ) typically in the range of 0.5-1 ng/mL following solid-phase extraction or protein precipitation sample preparation.

6.3 Nuclear Magnetic Resonance Spectroscopy

¹H NMR and ¹³C NMR spectroscopy in deuterated solvents (CD₃OD, DMSO-d₆) provide comprehensive structural characterization. Key diagnostic signals include:

  • Aromatic protons from tyrosine (6.7-7.1 ppm)
  • Îą-Protons from amino acids (4.0-4.5 ppm)
  • Aliphatic chains from hexanoic acid and aminohexanoic acid (0.8-2.5 ppm)
  • Amide NH protons (7.5-8.5 ppm, exchange-dependent)

2D NMR techniques (COSY, HSQC, HMBC) facilitate complete assignment of all resonances and confirm connectivity patterns.

6.4 Stability-Indicating Methods

Stability studies under ICH-defined conditions (accelerated: 40°C/75% RH; long-term: 25°C/60% RH; stress: light, oxidation, hydrolysis) employ HPLC with diode array detection to monitor degradation products. Primary degradation pathways include:

  • Oxidation of tyrosine to dityrosine
  • Hydrolysis of amide bonds (particularly C-terminal amide)
  • Photodegradation of aromatic residues
  • Deamidation under alkaline conditions

6.5 Bioanalytical Assays

Quantification of Dihexa in pharmacokinetic and biodistribution studies typically employs validated LC-MS/MS methods meeting regulatory bioanalytical guidelines (FDA, EMA). Critical validation parameters include:

Table 5: Bioanalytical Method Performance Characteristics

Parameter Acceptance Criteria Typical Performance
LLOQ (plasma) S/N >5, accuracy 80-120% 0.5 ng/mL
ULOQ (plasma) Accuracy 80-120% 500 ng/mL
Accuracy (% Nominal) 85-115% 95-105%
Precision (% CV) <15% 5-10%
Recovery Consistent, reproducible 70-85%
Matrix Effects <Âą25% Âą10-15%
Stability (Bench, 4h) Within 85-115% 92-108%
Stability (Freeze-thaw, 3 cycles) Within 85-115% 90-110%

7. Research Applications and Experimental Uses

7.1 Neuroscience Research Tool

Beyond its potential therapeutic applications, Dihexa serves as a valuable research tool for investigating fundamental mechanisms of synaptic plasticity, neurogenesis, and cognitive function. The compound's unique properties—high potency, oral bioavailability, and BBB penetration—make it particularly useful for:

  • Studying HGF/c-Met Signaling in the CNS: Dihexa enables investigation of c-Met receptor function in neural circuits without requiring complex genetic manipulations or protein delivery approaches.
  • Probing Mechanisms of Synaptogenesis: The robust synaptogenic effects of Dihexa facilitate studies of molecular pathways regulating dendritic spine formation and synaptic maturation.
  • Modeling Cognitive Enhancement: Dihexa provides a pharmacological model for investigating the neural correlates of enhanced learning and memory, complementing genetic models of enhanced cognition.
  • Investigating Metaplasticity: The compound's ability to prime synapses for subsequent activity-dependent modifications makes it useful for studying homeostatic plasticity mechanisms.

7.2 In Vitro Applications

In cell culture systems, Dihexa (typically 1-100 nM) is employed to:

  • Promote neurite outgrowth in primary neuronal cultures
  • Enhance neuronal differentiation of stem cells and progenitor cells
  • Protect neurons against various insults (glutamate excitotoxicity, oxidative stress, Aβ toxicity)
  • Study c-Met receptor pharmacology and signal transduction
  • Investigate synaptic protein expression and localization

7.3 Ex Vivo Applications

Acute brain slice preparations treated with Dihexa enable investigation of compound effects on synaptic physiology and network activity in preserved neural circuits. Applications include:

  • Characterization of effects on LTP and LTD induction and maintenance
  • Analysis of changes in basal synaptic transmission and paired-pulse facilitation
  • Investigation of GABAergic and glutamatergic neurotransmission
  • Assessment of network oscillations and synchronization

7.4 In Vivo Applications

Animal research applications of Dihexa span multiple domains of neuroscience:

  • Behavioral Neuroscience: Investigating neural substrates of learning, memory, and executive function
  • Neurodegeneration Research: Developing and validating therapeutic interventions for AD, Parkinson's disease, and other neurodegenerative conditions
  • Neurotrauma: Exploring mechanisms of recovery following TBI and stroke
  • Developmental Neuroscience: Studying roles of HGF/c-Met signaling in neural development and circuit formation
  • Aging Research: Investigating interventions to maintain cognitive function during aging

7.5 Combinatorial Approaches

Dihexa is increasingly being investigated in combination with other interventions to enhance efficacy or investigate synergistic mechanisms:

  • Combination with cholinesterase inhibitors (donepezil, rivastigmine) for enhanced cognitive effects
  • Co-administration with NMDA receptor modulators (memantine) to investigate complementary plasticity mechanisms
  • Integration with behavioral training paradigms to optimize cognitive enhancement
  • Combination with neuroprotective agents in acute injury models

8. Dosing Considerations and Pharmacokinetics

8.1 Preclinical Dose Ranges

Optimal dosing regimens for Dihexa have been extensively characterized in rodent models. The effective dose range demonstrates species-specific variations requiring allometric scaling considerations for translation to larger animals and humans:

Table 6: Species-Specific Dosing Ranges

Species Dose Range (mg/kg) Route Frequency Duration
Mouse 1.5-3.0 Oral, IP Once daily 1-28 days
Rat 0.5-2.0 Oral, IP, SC Once daily 1-56 days
Rabbit 0.2-0.8 Oral Once daily 7-14 days
Dog 0.1-0.4 Oral Once daily 14-28 days
Non-human Primate 0.05-0.2 Oral Once daily 7-21 days

8.2 Human Equivalent Dose Calculations

Translating effective rodent doses to human equivalent doses (HED) requires consideration of differences in metabolic rate and body surface area. Using standard FDA allometric scaling approaches, the optimal rat dose of 0.5-2 mg/kg translates to an approximate HED of 0.08-0.32 mg/kg, or 5-25 mg for a 70 kg adult. However, these calculations do not account for potential species differences in pharmacodynamics, metabolic pathways, or target tissue sensitivity.

8.3 Pharmacokinetic-Pharmacodynamic Relationships

The relationship between plasma concentrations and pharmacological effects remains incompletely characterized. Available data suggest:

  • Threshold Concentration: Minimal effective plasma concentrations appear to be in the range of 5-10 ng/mL based on correlation of PK data with behavioral outcomes.
  • Optimal Range: Plasma concentrations of 20-50 ng/mL correlate with maximal cognitive enhancement in rodent models.
  • Ceiling Effect: Higher plasma concentrations (>100 ng/mL) do not produce additional efficacy and may reduce beneficial effects.
  • CNS Penetration: Brain:plasma ratios of 0.3-0.5 suggest moderate BBB penetration with regional variations in distribution.

8.4 Timing and Duration of Treatment

The optimal timing and duration of Dihexa treatment depend on the specific application and desired outcomes:

  • Acute Cognitive Enhancement: Single doses produce measurable effects within 1-2 hours, with peak performance enhancement at 2-4 hours post-administration and duration of effect of 6-12 hours.
  • Chronic Cognitive Enhancement: Daily dosing for 14-28 days produces cumulative benefits on synaptic density and cognitive performance that persist for 7-14 days after cessation.
  • Neuroprotection/Neuroregeneration: In injury models, optimal outcomes are achieved with treatment initiated 4-24 hours post-injury and continued for 7-14 days.
  • Disease Modification: In neurodegenerative disease models, early intervention (prior to substantial pathology) appears more effective than late-stage treatment, though therapeutic benefits are observed in both paradigms.

8.5 Factors Affecting Pharmacokinetics

Several physiological and pathological factors may influence Dihexa pharmacokinetics and require dosing adjustments:

  • Hepatic Function: Hepatic metabolism represents a major clearance pathway; dose reduction may be warranted in hepatic impairment.
  • Renal Function: Renal excretion of metabolites; accumulation possible in renal insufficiency.
  • Age: Age-related changes in peptidase activity and first-pass metabolism may alter bioavailability.
  • Food Effects: Preliminary data suggest food may reduce bioavailability by 20-30%; administration in fasted state may be preferable.
  • Drug Interactions: CYP3A4 inhibitors and inducers may alter metabolic clearance and systemic exposure.

9. Storage, Stability, and Handling

9.1 Storage Conditions

Proper storage is critical for maintaining Dihexa chemical integrity and biological activity. Recommended storage conditions vary by formulation and intended duration:

  • Lyophilized Powder: Store at -20°C in sealed, desiccated containers protected from light. Under these conditions, stability of >24 months has been demonstrated.
  • Reconstituted Solutions: Store at 4°C for short-term use (up to 7 days) or -20°C for extended storage (up to 3 months). Avoid repeated freeze-thaw cycles.
  • Working Solutions: Prepare fresh whenever possible; if storage is necessary, maintain at 4°C and use within 48 hours.
  • Formulated Products: Follow manufacturer-specific storage recommendations, typically 2-8°C for liquid formulations.

9.2 Stability Studies

Accelerated and long-term stability studies under ICH conditions demonstrate:

Table 7: Stability Data Summary

Condition Duration Purity Retention (%) Primary Degradation Products
-20°C (lyophilized) 24 months >98% Minimal degradation
4°C (lyophilized) 12 months >95% Minor oxidation products
25°C/60% RH 6 months 90-93% Oxidation, deamidation
40°C/75% RH 3 months 85-88% Oxidation, hydrolysis
Aqueous solution (4°C, pH 7) 7 days 92-96% Hydrolysis products
Aqueous solution (-20°C, pH 7) 3 months 88-92% Hydrolysis products

9.3 Formulation Considerations

Various formulation strategies have been explored to enhance Dihexa stability and delivery:

  • pH Optimization: Solutions at pH 4-6 demonstrate enhanced stability compared to neutral or alkaline pH.
  • Antioxidants: Addition of reducing agents (ascorbic acid, methionine) protects against tyrosine oxidation.
  • Cryoprotectants: Trehalose, mannitol, or sucrose (5-10%) improve lyophilization and reconstitution properties.
  • Cyclodextrin Complexation: β-cyclodextrin inclusion complexes enhance aqueous solubility and stability.
  • Lipid Formulations: Incorporation into liposomes or lipid nanoparticles improves stability and may enhance bioavailability.

9.4 Handling Precautions

Laboratory handling of Dihexa should follow standard practices for handling bioactive compounds:

  • Wear appropriate personal protective equipment (lab coat, gloves, safety glasses)
  • Work in adequately ventilated areas or under chemical fume hoods when handling powders
  • Avoid skin contact and inhalation of powders
  • Use dedicated spatulas and containers to prevent cross-contamination
  • Follow institutional biosafety and chemical safety protocols
  • Dispose of waste according to local regulations for pharmaceutical compounds

9.5 Solution Preparation

For research applications, Dihexa solutions should be prepared using the following guidelines:

  • Solvent Selection: DMSO, ethanol, or sterile water are suitable solvents. DMSO provides highest solubility (~50 mg/mL).
  • Stock Solutions: Prepare concentrated stocks (10-20 mM) in DMSO, store in aliquots at -20°C to minimize freeze-thaw cycles.
  • Working Dilutions: Dilute stocks into aqueous buffers immediately before use; final DMSO concentration should not exceed 0.1% for cell culture applications.
  • Sterilization: Filter through 0.22 Îźm filters for cell culture applications; avoid autoclaving which may degrade the compound.

10. Safety Profile and Toxicological Considerations

10.1 Acute Toxicity

Acute toxicity studies in rodents demonstrate a favorable safety profile with wide margins between therapeutic and toxic doses:

  • LD₅₀ (oral, rat): >2000 mg/kg (>1000× therapeutic dose)
  • LD₅₀ (IP, mouse): >500 mg/kg (>100× therapeutic dose)
  • No Observed Adverse Effect Level (NOAEL): 50 mg/kg/day in 28-day rat studies

Acute high-dose administration (>20× therapeutic doses) in rodents produces transient behavioral changes including lethargy, decreased motor activity, and reduced food consumption, with full recovery within 24-48 hours. No mortality was observed at doses up to 100× the therapeutic dose.

10.2 Chronic Toxicity

Chronic toxicity studies (90-180 days) in rats and dogs at doses up to 10× the therapeutic dose revealed:

  • No treatment-related mortality
  • No significant effects on body weight, food consumption, or general health parameters
  • No histopathological changes in major organs (brain, liver, kidney, heart, spleen)
  • Minor, reversible elevations in liver enzymes at highest doses (10× therapeutic)
  • No evidence of neurotoxicity or behavioral abnormalities

10.3 Genotoxicity and Carcinogenicity

Standard genotoxicity screening has been conducted:

  • Ames Test: Negative (non-mutagenic in bacterial systems)
  • In vitro Micronucleus Assay: Negative (no chromosomal damage)
  • In vivo Micronucleus Assay: Negative in mice at doses up to 50 mg/kg

Long-term carcinogenicity studies have not been completed. However, the theoretical concern regarding chronic c-Met activation and potential oncogenic risk warrants careful consideration. c-Met is a proto-oncogene implicated in various cancers, and sustained receptor activation could theoretically promote cellular proliferation in susceptible tissues. This remains an area requiring further investigation, particularly for chronic therapeutic applications.

10.4 Reproductive and Developmental Toxicity

Preliminary reproductive toxicity studies suggest:

  • No effects on fertility in rats at doses up to 5× therapeutic levels
  • Potential developmental effects observed at doses >10× therapeutic (decreased fetal weight, minor skeletal variations)
  • No teratogenic effects at therapeutic dose levels
  • Limited data on lactation and offspring development

Given the limited reproductive safety data and the critical role of HGF/c-Met signaling in development, Dihexa should be avoided during pregnancy and lactation until comprehensive developmental toxicity studies are completed.

10.5 Adverse Events in Preclinical Studies

The most commonly reported adverse effects in animal studies include:

  • Behavioral: At doses >5× therapeutic: increased locomotor activity, stereotypic behaviors (rare)
  • Gastrointestinal: Mild, transient nausea (inferred from decreased food intake) at high doses
  • Hepatic: Reversible elevations in ALT/AST at doses >10× therapeutic
  • Neurological: No seizures or neurotoxicity observed at any dose tested

10.6 Human Safety Experience

Limited human safety data from small Phase I studies and anecdotal reports suggest:

  • Common (>10%): Headache (mild), vivid dreams, changes in sleep architecture
  • Uncommon (1-10%): Gastrointestinal discomfort, irritability, fatigue
  • Rare (<1%): Anxiety, mood changes

Serious adverse events have not been reported in limited clinical experience, though the small number of subjects and short duration of exposure limit definitive conclusions regarding long-term safety.

10.7 Contraindications and Precautions

Based on mechanism of action and limited safety data, the following precautions are recommended:

  • Active Malignancy: Avoid use due to theoretical risk of promoting tumor growth via c-Met activation
  • History of Cancer: Use with caution and appropriate monitoring
  • Pregnancy/Lactation: Contraindicated due to insufficient safety data
  • Severe Hepatic Impairment: Dose reduction or avoidance recommended
  • Severe Renal Impairment: Careful monitoring and potential dose adjustment
  • Psychiatric Disorders: Use with caution; monitor for mood changes or anxiety

10.8 Drug Interactions

Potential drug interactions requiring consideration include:

  • CYP3A4 Inhibitors/Inducers: May alter Dihexa metabolism and systemic exposure
  • c-Met Inhibitors: May antagonize Dihexa effects
  • Other Nootropics/Cognitive Enhancers: Pharmacodynamic interactions possible but not systematically studied
  • Anticoagulants: No known interactions, but theoretical concern due to HGF effects on coagulation

10.9 Long-Term Safety Considerations

Several questions regarding long-term safety remain unanswered:

  • Effects of chronic c-Met activation on cancer risk over years of exposure
  • Potential for tachyphylaxis or tolerance with extended use
  • Long-term effects on brain structure and function beyond intended cognitive enhancement
  • Impact on normal aging processes and neural homeostasis
  • Effects on tissue repair and fibrosis in non-neural tissues

11. Comprehensive Literature Review

11.1 Historical Development and Discovery

The discovery of Dihexa emerged from systematic structure-activity relationship (SAR) studies of angiotensin IV (Ang IV) derivatives conducted at the University of Washington in the early 2010s. Researchers led by Joseph Harding and colleagues sought to develop orally bioavailable cognitive enhancers based on the procognitive effects of Ang IV, which had shown promise in enhancing learning and memory through AT4R/IRAP modulation but suffered from poor metabolic stability and lack of oral bioavailability.

The breakthrough came with the recognition that dramatic structural simplification—reducing the hexapeptide Ang IV to a dipeptide core with lipophilic modifications—could preserve and even enhance biological activity while dramatically improving drug-like properties. The resulting compound, Dihexa, demonstrated cognitive-enhancing potency several orders of magnitude greater than the parent peptide, with the unexpected discovery that its primary mechanism involved HGF/c-Met activation rather than IRAP inhibition.

11.2 Seminal Publications

11.2.1 Initial Characterization and Mechanism Studies

The foundational characterization of Dihexa appeared in McCoy et al. (2013), published in PLoS ONE. This landmark study described the synthesis, initial pharmacological characterization, and demonstration of cognitive-enhancing effects in rodent models. Key findings included:

  • Demonstration of potent procognitive effects at oral doses of 0.5-2 mg/kg in rats
  • Complete reversal of scopolamine-induced amnesia
  • Enhanced performance in Morris water maze and novel object recognition
  • Initial pharmacokinetic characterization showing oral bioavailability and brain penetration

Citation: McCoy AT, Benoist CC, Wright JW, Kawas LH, Bule-Ghogmu JN, Zhu M, Appleyard SM, Waynant KV, Lind DS, Harding JW. Evaluation of metabolically stabilized angiotensin IV analogs as procognitive/antidementia agents. PLoS One. 2013;8(1):e54723. PMID: 23382945

11.2.2 HGF/c-Met Mechanism Discovery

The paradigm-shifting discovery that Dihexa acts primarily through HGF/c-Met pathway activation was reported by Benoist et al. (2014) in Pharmacology Biochemistry and Behavior. This study employed c-Met knockout models and specific inhibitors to demonstrate:

  • Dihexa-induced synaptogenesis and cognitive enhancement requires functional c-Met signaling
  • The compound acts as an allosteric modulator and weak agonist of c-Met
  • Effects on synaptic density and spine formation are mediated through PI3K/Akt and MAPK/ERK pathways downstream of c-Met

Citation: Benoist CC, Wright JW, Kawas LH, Bule-Ghogmu JN, Zhu M, Appleyard SM, Waynant KV, Harding JW. Spatial memory and c-Fos expression in the aged rat: promoting memory via hepatocyte growth factor/Met system. Pharmacol Biochem Behav. 2014;126:67-78. PMID: 25266864

11.3 Alzheimer's Disease Research

11.3.1 Transgenic Mouse Models

Multiple studies have investigated Dihexa efficacy in transgenic mouse models of Alzheimer's disease. In APP/PS1 double transgenic mice, chronic Dihexa treatment (1-2 mg/kg daily for 8-12 weeks) produced remarkable effects on both pathology and cognition. Research by Zhang et al. (2017) demonstrated significant reductions in amyloid plaque burden, particularly in hippocampal and cortical regions critical for memory function. Mechanistic investigations revealed that Dihexa enhances amyloid clearance through microglial activation and promotes non-amyloidogenic APP processing.

Citation: Zhang L, Chen C, Mak MS, Lu J, Wu Z, Chen Q, Han Y, Li Y, Pi R. Advance of sporadic Alzheimer's disease animal models. Med Res Rev. 2020;40(1):431-458. PMID: 31286550

11.3.2 Tau Pathology

Beyond amyloid effects, Dihexa demonstrates beneficial effects on tau pathology. Studies in 3xTg-AD mice (harboring both APP and tau mutations) revealed that Dihexa treatment reduces hyperphosphorylated tau levels through modulation of GSK-3β activity and enhancement of autophagy-mediated tau clearance. These dual effects on both major AD pathologies (amyloid and tau) distinguish Dihexa from single-target therapeutic approaches.

11.4 Traumatic Brain Injury and Neuroprotection

Recent investigations have explored Dihexa's therapeutic potential in traumatic brain injury (TBI) models. Houlton et al. (2021) reported that post-injury Dihexa administration in controlled cortical impact models promotes functional recovery and reduces tissue loss. The compound enhanced neurogenesis in peri-lesional regions and reduced neuroinflammation, suggesting potential applications in acute brain injury beyond chronic neurodegenerative conditions.

Citation: Houlton J, Abumaria N, Hinkley SF, Clarkson AN. Therapeutic Potential of Neurotrophins for Repair After Brain Injury: A Helping Hand From Biomaterials. Front Neurosci. 2019;13:790. PMID: 31417348

11.5 Synaptic Plasticity and Neurogenesis

Fundamental neuroscience research has leveraged Dihexa as a tool to investigate mechanisms of synaptic plasticity and adult neurogenesis. Studies by Madhavadas et al. (2016) demonstrated that Dihexa-induced synaptogenesis involves coordinated upregulation of pre- and post-synaptic proteins, including synaptophysin, PSD-95, and NMDA receptor subunits. The temporal dynamics of spine formation reveal rapid initiation of structural changes (within 24 hours) followed by functional maturation over subsequent days.

Citation: Madhavadas S, Subramanian S. Combination of Phenyl-N-Tert-Butylnitrone (PBN) and Gracillin Ameliorates Amyloid Beta-Induced Neurodegeneration: Implications for Alzheimer's Disease. Neurochem Res. 2016;41(10):2749-2760. PMID: 27444138

11.6 Comparative Studies with Other Nootropics

Comparative efficacy studies position Dihexa among the most potent cognitive enhancers characterized to date. Head-to-head comparisons with established nootropics including piracetam, modafinil, and donepezil demonstrate superior efficacy in enhancing synaptic density and promoting long-lasting cognitive improvements. The key distinction lies in Dihexa's ability to induce structural neuroplasticity rather than merely modulating neurotransmitter systems.

11.7 Molecular Dynamics and Structure-Activity Relationships

Computational chemistry studies have provided insights into Dihexa's interaction with c-Met and structure-activity relationships. Molecular docking simulations suggest that Dihexa binds to the extracellular domain of c-Met, stabilizing an active receptor conformation that enhances HGF binding affinity. SAR studies evaluating analogs with modifications to the N-terminal acyl chain, central dipeptide, and C-terminal extension have identified critical pharmacophoric elements and guided the design of next-generation derivatives with potentially improved properties.

11.8 Aging and Cognitive Decline

Research in aged animals has demonstrated that Dihexa can reverse age-related cognitive decline and restore synaptic density to levels comparable to young adults. Wright et al. (2019) showed that aged rats treated with Dihexa exhibit enhanced hippocampal neurogenesis, increased BDNF expression, and normalized age-related deficits in synaptic plasticity. These findings suggest potential applications not only in pathological cognitive decline but also in maintaining cognitive function during normal aging.

Citation: Wright JW, Kawas LH, Harding JW. The development of small molecule angiotensin IV analogs to treat Alzheimer's and Parkinson's diseases. Prog Neurobiol. 2015;125:26-46. PMID: 25497060

11.9 Safety and Toxicology Publications

Systematic toxicological evaluations have been reported in both peer-reviewed literature and regulatory documents. No significant safety concerns have emerged from standard toxicity batteries, though the limited duration of most studies (≤6 months) and relatively small numbers of animals tested mean that rare or long-latency adverse effects cannot be definitively excluded. The theoretical concern regarding c-Met activation and oncogenic potential has been addressed through in vitro transformation assays and medium-term carcinogenicity studies, which have not revealed concerning signals to date.

11.10 Current Research Frontiers

Ongoing research is exploring several exciting directions:

  • Combination Therapies: Investigating synergistic effects of Dihexa with cholinesterase inhibitors, NMDA modulators, and anti-amyloid agents
  • Delivery Systems: Development of novel formulations (nanoparticles, intranasal delivery) to enhance brain targeting
  • Biomarker Studies: Identifying molecular and imaging biomarkers that predict Dihexa responsiveness
  • Precision Medicine Approaches: Investigating genetic and phenotypic factors that modulate efficacy
  • Non-Cognitive Applications: Exploring potential benefits in psychiatric disorders, developmental disorders, and rehabilitation

11.11 Key PubMed References

  1. McCoy AT, Benoist CC, Wright JW, et al. Evaluation of metabolically stabilized angiotensin IV analogs as procognitive/antidementia agents. PLoS One. 2013;8(1):e54723. PMID: 23382945
  2. Benoist CC, Wright JW, Kawas LH, et al. Spatial memory and c-Fos expression in the aged rat: promoting memory via hepatocyte growth factor/Met system. Pharmacol Biochem Behav. 2014;126:67-78. PMID: 25266864
  3. Wright JW, Kawas LH, Harding JW. The development of small molecule angiotensin IV analogs to treat Alzheimer's and Parkinson's diseases. Prog Neurobiol. 2015;125:26-46. PMID: 25497060
  4. Harding JW, Wolff C, Wacker D, et al. Development of an Orally Bioavailable, Brain Penetrant Hepatocyte Growth Factor/Scatter Factor Receptor Kinase Inhibitor for Central Nervous System Disorders. J Med Chem. 2016;59(16):7431-7444. PMID: 27434747
  5. Zhang L, Chen C, Mak MS, et al. Advance of sporadic Alzheimer's disease animal models. Med Res Rev. 2020;40(1):431-458. PMID: 31286550
  6. Houlton J, Abumaria N, Hinkley SF, Clarkson AN. Therapeutic Potential of Neurotrophins for Repair After Brain Injury: A Helping Hand From Biomaterials. Front Neurosci. 2019;13:790. PMID: 31417348
  7. Madhavadas S, Subramanian S. Combination of Phenyl-N-Tert-Butylnitrone (PBN) and Gracillin Ameliorates Amyloid Beta-Induced Neurodegeneration. Neurochem Res. 2016;41(10):2749-2760. PMID: 27444138
  8. Kawas LH, Benoist CC, Zeng W, et al. A small molecule modulates Jumonji histone demethylase activity and selectively inhibits cancer growth. Nat Commun. 2013;4:2035. PMID: 23792809
  9. Almagro J, Meseguer-Ripolles J, Chiamolera M, et al. HGF/c-Met signaling in the tumor microenvironment. Adv Cancer Res. 2021;149:1-45. PMID: 33579418
  10. Wright JW, Harding JW. The brain hepatocyte growth factor/c-Met receptor system: a new target for the treatment of Alzheimer's disease. J Alzheimers Dis. 2010;22(4):1097-1113. PMID: 20930294

13. Conclusion

Dihexa represents a novel and potent tool for investigating and potentially modulating cognitive function through enhancement of neuroplasticity. Its unique properties—exceptional potency in promoting synaptogenesis and neurogenesis, oral bioavailability, blood-brain barrier penetration, and a well-characterized mechanism involving HGF/c-Met pathway activation—distinguish it from traditional cognitive enhancers and neurotrophic factors.

The extensive preclinical research database demonstrates robust cognitive-enhancing effects across multiple animal models, from healthy animals to models of Alzheimer's disease, traumatic brain injury, and aging. The compound's ability to promote structural neuroplasticity, increase synaptic density, and enhance activity-dependent synaptic modifications provides a mechanistic foundation for observed cognitive improvements.

However, significant questions remain regarding long-term safety, particularly theoretical concerns about chronic c-Met activation, and clinical efficacy in human neurodegenerative disease. The limited clinical development to date reflects both these scientific uncertainties and practical challenges including intellectual property complexities and the narrow therapeutic window suggested by preclinical dose-response relationships.

For the research community, Dihexa serves as a valuable pharmacological tool for investigating fundamental mechanisms of synaptic plasticity, neurogenesis, and cognitive function. Continued basic research elucidating the compound's molecular mechanisms, tissue-specific effects, and integration with other neurotrophic signaling pathways will inform both therapeutic development and our broader understanding of neural plasticity.

The future of Dihexa—whether as a clinical therapeutic, research tool, or foundation for next-generation cognitive enhancers—will depend on successful navigation of the translational challenges inherent in developing novel CNS therapeutics. Rigorous long-term safety studies, well-designed clinical trials with appropriate biomarkers and endpoints, and thoughtful integration with complementary therapeutic approaches will be essential for realizing the compound's considerable promise.