miércoles, 23 de agosto de 2023

Heroin and naltrexone effects on pituitary-gonadal hormones in man: interaction of steroid feedback effects, tolerance and supersensitivity.

 J H Mendelson, J Ellingboe, J C Kuehnle and N K Mello. Journal of Pharmacology and Experimental Therapeutics September 1980,

Abstract

The acute and chronic effects of heroin, and the opiate antagonist naltrexone, on integrated plasma samples analyzed for luteinizing hormone (LH) and testosterone (T) levels were studied in six adult males with a history of heroin addiction. Acute doses of heroin (10 mg i.v.) significantly suppressed LH levels. Chronic heroin use was also associated with a significant decrease in plasma T levels. LH levels after chronic heroin use were lower than control levels but the degree of LH suppression was approximately the same as after the acute doses of heroin. Acute naltrexone administration did not alter T levels appreciably but was associated with a significant elevation in LH levels. After 22 days of chronic naltrexone maintenance, T levels were in the high normal range and LH levels were in the low normal range. These data suggest the development of tolerance and supersensitivity to opiate agonist and antagonist effects on pituitary-gonadal hormones involves interaction between the direct effects of these drugs on LH followed by steroid feedback effects of T on gonadotrophin secretory activity.

Antitumor activity of naltrexone and correlation with steroid hormone receptors

 Abstract

We have evaluated the opiate peptide antagonist, naltrexone, for its effectiveness as an antitumor agent. For this evaluation, we tested the effect of naltrexone given daily in the diet on the growth of established 7,12-dimethylbenz(a)anthracene-induced mammary tumors. Tumors continued to grow actively in rats fed chow diet only (control group). In contrast, the naltrexone-supplemented diet (75 mg/kg diet) significantly decreased the size of the established mammary tumors in rats over the 25 day observation period, resulting in an average decrease in tumor volume by approximately 23% compared with their sizes at the beginning of the treatment. Tumor regression occurred in 70% of the rats. Tumors that respond to naltrexone showed appreciable amounts of estrogen and progesterone receptors while unresponsive tumors were negative for estrogen and progesterone receptors. For the first time, we report that naltrexone can regress established mammary tumors and that the inhibitory effect of naltrexone appears to be restricted to the hormonally responsive mammary tumors.

lunes, 7 de agosto de 2023

Influence of low dose naltrexone on Raman assisted bone quality, skeletal advanced glycation end-products and nano-mechanical properties in type 2 diabetic mice bone

 © 2021 - Abstract

Type 2 diabetes mellitus (T2DM) commonly affects the bone mineral phase and advanced glycation end-products (AGEs) which eventually led to changes in bone material properties on the nano and macro-scale. Several anti-diabetic compounds are widely used to control high blood sugar or glucose caused by T2DM. 

Low Dose Naltrexone (LDN), an opiate receptor antagonist, and a known TLR4 antagonist, treatment can improve glucose tolerance and insulin sensitivity in high-fat-diet (HFD) induced T2DM mice. However, the influences of LDN on the local bone quality, mineralization of the bone, and the skeletal AGEs levels have not been fully elucidated. The objective of this study is to understand the effect of LDN on Raman assisted bone quality, skeletal AGEs (determined by Raman spectroscopy), and nano-mechanical properties in HFD induced T2DM mice bone. In order to investigate these, mice and corresponding bones were divided into four groups (divided based on diet and treatment), (a) normal control diet treated with saline water, (b) normal control diet treated with LDN, (c) HFD treated with saline water, and (d) HFD treated with LDN. In T2DM condition (HFD treated with saline water), alteration of Raman-based compositional measures in bone quality including mineral-to-matrix ratios, carbonate substitution, mineral crystallinity, and collagen quality was observed. Our data also indicated that T2DM enhances the skeletal AGEs, and impairs the nano-mechanical properties. 

Interestingly, present results indicated that LDN controls the Raman-based compositional measures in bone quality in HFD induced T2DM mice bone. Additionally, LDN also protects the alteration of the skeletal AGEs levels and nano-mechanical properties in T2DM mice bone. This study concluded that LDN can control the HFD induced T2DM affected bone abnormalities at multiple hierarchical levels.


Keywords: Bone; Mineralization; Naltrexone; Nano properties; Raman spectroscopy; T2DM.

LDN Protects Bone Property Deterioration at Different Hierarchical Levels in T2DM Mice Bone

 2021 Aug 10. - Type 2 diabetes mellitus (T2DM) commonly affects bone quality at different hierarchical levels and leads to an increase in the risk of bone fracture. Earlier, some anti-diabetic drugs showed positive effects on bone mechanical properties. Recently, we have investigated that low-dose naltrexone (LDN), a TLR4 antagonist treatment, improves glucose tolerance in high-fat diet (HFD)-induced T2DM mice and also gives protection against HFD-induced weight gain. However, effects on bone are still unknown. In this study, the effects of LDN on the bone properties at different hierarchical levels in T2DM mice bone were investigated. In order to investigate these, four different groups of bone (divided based on diet and treatment) were considered in this present study. These are (a) normal control diet treated with saline water, (b) normal control diet treated with LDN, (c) HFD treated with saline water, and (d) HFD treated with LDN. Bone properties were measured in terms of fracture toughness, nano-Young's modulus, hardness, mineral crystal size, bone composition, and bulk mineral to matrix ratio. Results indicated that fracture toughness, nano-Young's modulus, and hardness were decreased in T2DM bone as compared to normal bone, and interestingly, treatment with the LDN increases these material properties in T2DM mice bone. Similarly, as compared to the normal bone, decrease in the mineral crystal size and bulk mineral-to-matrix ratio was observed in the T2DM bone, whereas LDN treatment protects these alterations in the T2DM mice bone. The bone size (bone geometry) was increased in the case of HFD-induced T2DM bone; however, LDN cannot protect to increase the bone size in the T2DM mice bone. 

In conclusion, LDN can be used to control the T2DM-affected bone properties at different hierarchical levels.


Acute Low Dose Naltrexone Increases β-Endorphin and Promotes Neuronal Recovery Following Hypoxia-Ischemic Stroke in Type-2 Diabetic Mice

 Abstract  -   2023 May 11.

Diabetic patients experience significant mortality and poor recovery following ischemic stroke. Our clinical and basic science studies demonstrate an overall immune suppression in the periphery of diabetic stroke patients, as well as within the central nervous system (CNS) of type-2 diabetic mice following hypoxia-ischemia (HI). Low doses of naltrexone (LDN) improved clinical outcomes in many autoimmune diseases by acting on opioid receptors to release β-endorphin which in turn balances inflammatory cytokines and modulates the opioid growth factor (OGF)-opioid growth factor receptor (OGFr) pathway. We hypothesized that in our model of diabetic mice, LDN treatment will induce the release of β-endorphin and improve CNS response by promoting neuronal recovery post HI. To test this hypothesis, we induced HI in 10 week old male db/db and db/ + mice, collected tissue at 24 and 72 h post HI, and measured OGF levels in plasma and brain tissue. The infarct size and number of OGF + neurons in the motor cortex, caudate and hippocampus (CA3) were measured. Following HI, db/db mice had significant increases in brain OGF expression, increased infarct size and neurological deficits, and loss of OGFr + neurons in several different brain regions. In the second experiment, we injected LDN (1 mg/kg) intraperitoneally into db/db and db/ + mice at 4, 24, and 48 h post HI, and collected brain tissue and blood at 72 h. Acute LDN treatment increased β-endorphin and OGF levels in plasma and promoted neuronal recovery in db/db mice compared to phosphate buffer saline (PBS)-treated diabetic mice suggesting a protective or regenerative effect of LDN.


Keywords: Diabetes; Hypoxia-ischemia; Naltrexone; Opioid growth factor; β-endorphin.

Off-Label, Low-Dose Naltrexone for Refractory Painful Diabetic Neuropathy

Published: 25 November 2015 

Here, we report a case in which off-label LDN was used for the treatment of painful diabetic neuropathic pain refractory to most available therapy.


In April 2012, a 76-year-old male with a 30-year history of type-2 diabetes and 7 years of diabetic neuropathic symptoms presented in the endocrinology clinic with complaints of burning pain in both legs below the mid-calf level. He described that the pain began insidiously, occurring off and on and associated with certain degree of numbness. Subsequently, both the frequency of occurrence and intensity of pain were increased disturbing his night-time sleep. The pain was partially relieved by walking, massage, hot fomentation, or paracetamol.


In July 2010, for the first time the patient sought treatment for neuropathic pain. He received Amitriptyline, Pregabalin, Duloxetine, Lamotrigine, and nonsteroidal anti-inflammatory drugs (NSAIDs) in varying doses and combinations. All drugs and combinations were tried for at least 1–2 months. Subsequently, he underwent a lumbar paravertebral nerve block (L2-L4) and had near complete pain relief afterward but the pain reappeared in a few weeks. He also received injectable vitamin B-complex and vitamin-D in therapeutic doses without any benefit. Opiates, transdermal patches of capsaicin, or lidocaine were not used.


On examination of the lower limbs, the skin over the dorsum of both the feet was shiny without any ulcer suggesting good foot care. The touch perception was decreased bilaterally below the knees and there was hyperalgesia, but no allodynia. The temperature sensation was normal in both legs. There was decreased vibration perception on both feet. The joint position sensation was lost in all joints on both feet but preserved in knees and above. The patient rated his pain to be 90% (0–100 points), 8, and 8 on a visual analog scale (VAS), short form McGill pain questionnaire, and 11-point Likert pain scale, respectively. The neuropathy symptom score was 9 out of 9. The deep tendon reflexes were absent in both ankles, 1+ in both knees, and 2+ in the joints of the upper limbs. Muscle power was normal in all limbs. Nerve conduction studies showed bilateral sensory motor polyneuropathy. The patient had adequate glycaemic control (HbA1c, 6.4%) and was on metformin (2 g/day), pioglitazone (30 mg/day), and insulin (20 units/day; 30% soluble and 70% isophane) with good compliance. Workup for other causes of neuropathy was non-contributory. MRI of lumbar spine showed degenerative changes without any neural involvement.


Based upon earlier reports [ 4 ], it was planned to administer oral naltrexone in graded doses (1, 2, and 4 mg HS for 2 weeks each). The 1 mg dose did not show any appreciable response. However, with the 2 mg dose, the patient reported a partial improvement in the burning pain. The 4 mg dose for 2 weeks produced a much greater pain relief. He rated his pain to be 5% on VAS as compared to 90% before therapy. On the Likert scale for pain and short-form McGill pain questionnaire the scores reduced to 1 and 2, respectively. Sleep was good after the treatment. On examination, there was no hyperalgesia, but the sensory loss was not improved. Following naltrexone therapy, initially he experienced mild diarrhea, nausea, and somnolence, which subsided spontaneously in a few days without any intervention. At every follow up the patient was satisfied with LDN (4 mg HS) and was continuing the same dose until October 2014 (last follow up) without experiencing any significant side effect.


The proposed mechanisms of pain relief with LDN include opioid receptor blockade causing compensatory release of endogenous opioids, and antagonism of Toll-like receptor-4 on microglia, which produces a variety of inflammatory factors such as pro-inflammatory cytokines, substance-P, nitric oxide, and excitatory amino acids [ 5,6 ]. Other proposed targets include astrocytes [ 7 ] NADPH oxidase-2 [ 8 ], and opioid growth factor receptor (OGFr) [ 9 ]. So far, very little information is available on the mechanism of pain relief by LDN and its long-term safety.


To our knowledge, this is the first report demonstrating the efficacy of LDN in relieving the pain of diabetic neuropathy. Based upon the present findings, we feel the need for further research to elucidate the possible mechanism(s) of LDN focussing on the roles of endogenous opioids and neuroinflammation, and to conduct large randomized, double-blind, clinical trials to establish the possible mechanism, efficacy, and safety of LDN in painful diabetic neuropathy and other chronic painful conditions.


Efficacy and safety of low-dose naltrexone in painful diabetic neuropathy: A randomized, double-blind, active-control, crossover clinical trial

 jun 2021 - Background: There is a need for newer therapies for chronic painful diabetic neuropathy as the existing drugs have their own limitations. Clinical trials on low-dose naltrexone (1-5 mg/d) showed efficacy and safety in certain chronic painful conditions, but not in painful diabetic neuropathy. Hence the present study was planned.


Methods: Sixty-seven participants with painful diabetic neuropathy were randomized to receive either 2 mg naltrexone or 10 mg amitriptyline daily following a 2-week run-in period. The participants were followed up every 2 weeks for a total of 6 weeks. Up-titration was done (to 4 mg naltrexone or 25/50 mg amitriptyline) if the pain reduction was less than 20% on the visual analog scale (VAS) during the next follow-up visit. Efficacy was assessed using the change in VAS score at the end of 6 weeks from baseline. Safety was evaluated at each follow-up visit. After 2 weeks of washout period, the participants were crossed over to receive the comparator drug for another 6 weeks with similar evaluations.


Results: The difference (confidence interval) in the change in VAS score between groups from baseline was 1.64 (-0.92 to 4.20) in per-protocol analysis and 1.5 (-1.11 to 4.13) in intention-to-treat analysis. Eight and fifty-two adverse events were reported in the naltrexone and amitriptyline groups, respectively (P < .001). The most common adverse events were mild diarrhea with naltrexone and somnolence with amitriptyline.


Conclusions: Low-dose naltrexone exhibited similar efficacy and a superior safety profile compared with amitriptyline in painful diabetic neuropathy.


Keywords: amitriptyline; clinical trial; diabetes; low-dose naltrexone; painful neuropathy

Influence of chronic naltrexone treatment on growth hormone secretion in normal subjects

1997 Dec; 

Abstract

Objective: To verify if a chronic opioid blockade could affect the GH/IGF-I axis.

Design: We have investigated the effects of naltrexone (NTX) treatment on GH response to GHRH in normal women.

Methods: GHRH test (50 micrograms i.v.) performed in seven normal female volunteers (age 25-38 years, with a body mass index ranging from 19.8 to 23.1 kg/m2) before and after 4-weeks NTX treatment (50 mg p.o. daily).

Results: Basal GH, IGF-I, insulin-like growth factor binding protein-3 (IGFBP-3) plasma levels and the IGF-I/IGFBP-3 molar ratio remained unaffected by NTX. NTX significantly reduced the GH peak values (15.52 +/- 3.59 vs 4.78 +/- 0.49 micrograms/l; P < 0.01), and GH area under curve (918.93 +/- 253.96 vs 401.09 +/- 79.63 micrograms/l; P < 0.01).


Conclusions: This finding suggests that the long-term opioid receptor blockade has an inhibitory role on GHRH-induced GH secretion. A central influence on neurotransmitter control of GH might be hypothesised. The inhibition of stimulated GH release, without interference with the basal level, could indicate an enhanced somatostatin secretion and/or activity. Opioids could be involved only in the regulation of GH dynamics and not in basal secretion. Nevertheless, a direct involvement of opioids at the pituitary level, which could be modified by NTX, cannot be excluded. 

sábado, 5 de agosto de 2023

Increase Growth Hormone with Naltrexone

Naltrexone, according to certain studies, has shown to help increase growth hormone levels in the body. Growth hormone is directly responsible for maintaining and building lean muscle mass. It also helps in burning fat that contributes to increased weight loss.

It is a known fact that growth hormones start decreasing as your weight increases. Growth hormones are thought to be an indicator of weight loss or gain along with calorie consumption. The relationship between insulin and growth hormones is an interesting concept as well.

Growth hormones decrease as insulin production increases in the body. Therefore, the more you gain, the lower your growth hormone levels will be. This will contribute to an increase in insulin. Low dose naltrexone is beneficial in decreasing insulin levels that helps in improving growth hormone levels.

The bottom line is that naltrexone can increase growth hormone levels in obese people that may improve metabolism and lean muscle mass.

Naltrexone treatment restores menstrual cycles in patients with weight loss-related amenorrhea

Plasma gonadal steroid levels increased in all patients and in 24 of 30 patients the menstrual bleeding occurred within 90 days from the beginning of treatment. After 6 months from naltrexone discontinuation, 18 of 24 patients still showed the occurrence of menstrual cycles. Luteinizing hormone plasma levels and LH pulse amplitude increased after 3 months of treatment and remained unchanged 6 months after naltrexone suspension. Plasma FSH levels did not show any change in any patient. The body mass index increased after 3 months in all patients who menstruated. Patients treated with placebo did not show any significant change in gonadotropins and gonadal steroid plasma levels.

Hydrogels for diabetic eyes: Naltrexone loading, release profiles and cornea penetration

 Abstract

Naltrexone (NTX) is a potent opioid growth factor receptor (OGFR) antagonist proved to be useful for treatment of ocular surface complications. The aim of this work was to explore the feasibility of designing NTX-imprinted 2-hydroxyethyl methacrylate-based hydrogels for sustained drug release on the ocular surface. Acrylic acid (AAc) and benzyl methacrylate (BzMA) were chosen as functional monomers able to form binding cavities mimicking OGFR binding sites for NTX. Imprinted hydrogels containing functional monomers loaded higher amounts of NTX compared to non-imprinted ones by simple soaking in drug aqueous solution. In addition, possibility of carrying out the loading and sterilization processes in one step was investigated. NTX release was evaluated both under agitated sink conditions and in a microfluidic flow chamber mimicking the hydrodynamic conditions of the eye, namely the small volume of lachrymal fluid and its renovation rate. Sustained release profiles together with adequate swelling degree (46 to 57% w/w), light transparency (over 85%) and oxygen permeability may make these hydrogels suitable candidates to NTX-eluting contact lenses. NTX-loaded and non-loaded discs successfully passed the chorioallantoic membrane test for potential ocular irritation and were cytocompatible with human mesenchymal stem cells. Finally, NTX-imprinted hydrogels tested in the bovine corneal permeability assay provided therapeutically relevant amounts of NTX inside the cornea, reaching drug levels similar to those attained with a concentrated aqueous solution in spite the discs showed sustained release.


Keywords

Naltrexone Contact lenses Imprinted hydrogels Diabetic eye complications Corneal diseases Dry eye

1. Introduction

Cornea, sclera, conjunctiva and tear film are functionally linked as one system, including tear glands, to protect the eye against external adverse events and pathogens [1]. In addition, lachrymal fluid and cornea are the first refracting elements. Thus, cornea integrity and an adequate tear production are essential to preserve a healthy vision [2]. Metabolic changes suffered by persons with diabetes lead to important ocular surface complications such as decreased tear production, diminished corneal sensitivity, and delayed wound healing [3]. Disruption of tear film barrier function and poor tear quality production can also result in irritation, inflammation of the stroma and impaired vision.


Enkephaline is a natural opioid growth factor (OGF) that, when interacts with its receptor (OGFr), negatively regulates cell proliferation and tissue growth. Enkephaline has been found to be at higher levels in persons with diabetes compared to healthy people [4,5]. Elevated concentrations of this peptide, which have also been confirmed in diabetes animal models, are responsible for complications such as ulceration and increased susceptibility to infection due to delayed epithelialization during wound healing. It should be noted that the OGF-OGFr axis plays a key role in the homeostasis of cornea and retina [1], and thus antagonists of OGFr, such as naltrexone (NTX), may revert ocular surface complications by blocking the effects of enkephaline [4,6]. NTX is approved in oral formulations for treatment of addiction to opioids or alcohol, although it undergoes a relevant hepatic first pass effect. Preclinical studies have demonstrated that NTX, either systemically or topically applied, can notably improve corneal wound healing and reverse severe dry eye [6,7]; for example, studies on type I diabetic rats and normal rats with episodic dry eye have shown that one drop of NTX restore tear production in <1 h and the effect persists for 72 h [6]. Importantly, repeated doses of NTX eye drops (50 μM; 4 drops) over a 24 h period did not cause adverse events on healthy human volunteers [2]. However, NTX tends to autoxidation when it is formulated as eye drops [8]. Furthermore, conventional ocular formulations provide low ocular bioavailability because of short precorneal residence time and limited cornea permeability. Thus, to achieve a therapeutic effect, frequent instillations are needed. Alternatively, mucoadhesive in situ gelling formulations [9] and niosomes [10] have been explored.


Contact lenses (CLs) are safe devices widely used to correct refractive errors and are attracting an increasing interest as drug delivery platforms [[11], [12], [13], [14]]. Several approaches have been implemented to endow CLs with ability to host drugs such as timolol, acetazolamide, olopatadine, amphotericin B, ciprofloxacin or epalrestat, among others [[15], [16], [17], [18], [19], [20]]. CLs can act as a reservoir that provides sustained levels of drug in the precorneal area (postlens lachrymal fluid) while minimizes drug loss due to blinking, reflex lachrymation or nasolacrimal drainage [21]. One useful approach to enhance the capability of CLs to load therapeutic amounts of drug is the molecular imprinting [11,22]. This technique relies on the use of the target drug molecules as templates during polymerization in order to induce monomers arrangement as a function of their affinity. After polymerization, the template molecules are removed and specific cavities for the target drug, named imprinted pockets, are revealed. Physical stability of the imprinted cavities is quite limited in the case of loosely crosslinked networks, as occurs for soft CLs. Therefore, a precise selection of functional monomers that can endow the cavities with sufficient affinity for the target molecule even after the swelling of the network is required for the success of the recognition. Bioinspired strategies that rely on mimicking the pharmacological target of the drug in the hydrogel network have been shown useful for the rational selection of functional monomers [17,23,24].


The aim of the present work was to design NTX-imprinted poly(2-hydroxyethyl methacrylate) (pHEMA) hydrogels suitable as soft CLs that can load therapeutic amounts of NTX and provide sustained release on the ocular surface. The hydrogels were designed taking into account the information available about the interactions of NTX with the μ-opioid receptor (MOR) [25,26], which are mainly driven by binding to amino acid residues Asp147 (bearing a carboxylic acid group) and Tyr148 (bearing a phenyl group) and polar interactions with Lys233 [27,28]. Taking into account this information, functional monomers were chosen among those bearing carboxylic acid groups (acrylic acid, AAc) and aromatic groups (benzyl methacrylate, BzMA). It can be hypothesized that the addition of NTX before polymerization may drive the adequate spatial arrangement of the monomers for a more efficient formation of ad hoc artificial receptors (imprinted hydrogels). Both imprinted and non-imprinted hydrogels were carefully washed and then loaded with NTX by soaking in aqueous solutions. Feasibility of carrying out loading and sterilization processes in one step was investigated. Since there is not a standardized release test for ocular solid or semisolid formulations, the diversity of setups and conditions reported in literature (some far from mimicking cornea environment) make comparisons and predictions difficult [29]. Thus, to gain an insight into the effect of the tests conditions on the release profiles, NTX release from the hydrogels was recorded using both a conventional test in bulk medium and also a microfluidic device mimicking lachrymal fluid turn over [30] and the information obtained compared to elucidate whether correlations among both methods could be established. Finally, after confirming biocompatibility, most promising formulations were also evaluated regarding corneal accumulation and permeability.


2. Experimental

2.1. Materials

Naltrexone hydrochloride (NTX), ethylene glycol dimethacrylate (EGDMA) and dichlorodimethylsilane were from Sigma-Aldrich (Steinheim, Germany); 2-hydroxyethyl methacrylate (HEMA) and acrylic acid (AAc) were from Merk (Dramstad, Germany); benzyl methacrylate (BzMA) from Polysciences Inc. (Warrington, UK) and 2,2′-azobis(2-methylisopropionitrile) (AIBN) from Across (New Jersey, USA). WST-1 cell proliferation reagent was from Roche (Mannheim, Germany); phosphate buffer saline (PBS), MEM Alpha and fetal bovine serum (FBS) were from Sigma-Aldrich (Saint Louis, USA); penicillin/streptomycin (10,000 U/mL and 10,000 μg/mL), l-glutamine (200 mM) and 0.25% trypsin-EDTA were from Gibco (Paisley, UK). Water was purified using reverse osmosis (resistivity > 18 MΩ·cm, MilliQ, Millipore® Spain). Simulated lachrymal fluid (SLF) was prepared with the following composition: 6.78 g/L NaCl from Scharlau (Barcelona, Spain), 2.18 g/L NaHCO3 from Panreac (Barcelona, Spain), 1.38 g/L KCl, and 0.084 g/L CaCl2·2H2O from Merck (Dramstadt, Germany) with pH 7.8. Carbonate buffer pH 7.2 was prepared mixing buffer solution A (100 mL): 1.24 g NaCl from Scharlau (Barcelona, Spain), 0.071 g KCl from Merck (Dramstadt, Germany), 0.02 g NaH2PO4·H2O from Merk (Dramstadt, Germany) and 0.49 g NaHCO3 from Panreac (Barcelona, Spain); and buffer solution B (100 mL): 0.023 g CaCl2 from Merck (Dramstadt, Germany) and 0.031 g MgCl2·6H2O from Scharlau (Barcelona, Spain).



2.8. HET-CAM test

The Hen's Egg Test on Chorio-Allantoic Membrane (HET-CAM) assay is a validated alternative to animal testing of ocular irritancy [34]. The HET-CAM test was carried out using fertilized hen's eggs (50–60 g; Coren, Spain) incubated at 37 °C and 60% RH during 7 days [34]. A rotator saw (Dremel 300, Breda, The Netherlands) was used to make a circular cut of 1 cm in diameter on the wider extreme (where the air chamber is placed), on the eighth day, to remove the eggshell. The inner membrane was wet with 0.9% NaCl for 30 min and then carefully removed to expose the chorioallantoic membrane (CAM). NTX aqueous solution (0.3 mg/mL, 300 μL) and hydrated drug-loaded discs (imprinted and non-imprinted in duplicate) were placed on the CAM. 0.9% NaCl and 0.1 N NaOH solutions (300 μL) were used in triplicate as negative and positive controls, respectively. The vessels of CAM were observed during 5 min, under white light, for haemorrhage, vascular lysis or coagulation. The irritation score (IS) was calculated as previously reported [20,35].


2.9. Cytocompatibility assay

Human mesenchymal stem cells derived from bone marrow (hMSC, ATCC-PCS-500-012™) were cultured in 175 cm2 culture flask with MEM Alpha supplemented with fetal bovine serum heat inactivated (FBS, 10%), and antibiotics (penicillin/streptomycin 10,000 Units/mL and 10,000 μg/mL respectively, 1%). hMSCs were harvested, at approximately 90% of confluence, with 0.25% trypsin-EDTA. Cells were seeded (10,000 cells/well) in 24 wells plate and kept at 37 °C, 5% CO2 and 95% HR for 12 h allowing cell adhesion.


Pieces of NTX-loaded and non-loaded imprinted hydrogels bearing AAc and BzMA (19.6 mm2) were placed in the 24 wells plate (10,000 cells/well) the plate was kept at 37 °C, 5% CO2 and 95% HR. Solution of NTX (50 μM) in MEM Alpha was used as control. After 72 h, WST-1 cell proliferation assay was carried out following manufacturer instructions. Briefly, the medium was replaced by MEM Alpha (0.5 mL, with no supplements), and WST-1 cell proliferation reagent (50 μL; Roche, Mannheim, Germany) was added. Plates were incubated at 37 °C, 5% CO2 and 95% HR for 1 h and measured spectrophotometrically at 450 nm in a Bio-Rad plate reader 680 (California, USA).


2.10. Bovine corneal permeability test

Fresh bovine eyeballs were collected from the local slaughterhouse and transported following BCOP protocol alternative to in vivo testing [36]. Eyes were carried completely immersed in PBS with antibiotics 1% (100 IU/mL penicillin, 100 μg/mL streptomycin) in an ice bath. Once arrived, corneas were excised with 2–3 mm of surrounding sclera, rinsed with PBS and mounted in vertical diffusion (Franz) cells. Carbonate buffer pH 7.2 (6 mL) was used to fill receptor chamber and a small stir bar was also incorporated. The cornea was placed on the receptor chamber (maintained at 37 °C) and the donor chamber was fixed and filled also with carbonate buffer pH 7.2 (0.785 cm2 area available for permeation). After 1 h equilibration, the buffer in the donor chamber was removed and the corneas were exposed to NTX-loaded discs (C2 and D2 discs with 1 mL of 0.9% NaCl) or to control drug solution (350 μg/mL NTX, 2 mL). All experiments were carried out in triplicate. The donor chamber was covered with parafilm to prevent evaporation. Samples (1 mL) were taken from the receptor chamber at 1, 2, 3, 4, 5 and 6 h, replacing with the same volume of bicarbonate buffer each time and taking care of removing air bubbles from the diffusion cell. NTX in the receptor medium was quantified by means of an HPLC equipment (Waters 717 Autosampler, Waters 600 Controller, 996 Photodiode Array Detector) fitted with a C18 column (Waters Symmetry C18 5 μm; 3.9 × 150 mm) and Empower2 software. Mobile phase consisted of acetonitrile: 10 mM ammonium acetate buffer (40:60 v/v, pH adjusted to 5.6 with acetic acid) at 0.5 mL/min and 30 °C [37]. The injection volume was 50 μL, and naltrexone was quantified at 220 nm (retention time 3.08 min). Calibration was carried out with NTX standard solutions (1–10 μg/mL) in water (filtered through 0.2 μm, 13 mm GMP Minispike filters).


After 6 h of test, the formulations were removed from the donor chambers and the corneas were rinsed at least three times with PBS. Then, the corneas were placed in tubes with 2 mL acetonitrile for 24 h. The amounts of NTX extracted from the cornea were measured by HPLC as explained above.


2.11. Statistical analysis

The effects of sterilization process on NTX loading, and of hydrogels with and without NTX on NTX diffusion coefficients, hMSCs viability and cornea permeability were analyzed using ANOVA and multiple range test (Statgraphics Centurion XVI 1.15, StatPoint Technologies Inc., Warrenton VA).


3. Results and discussion

3.1. Hydrogel synthesis

Naltrexone hydrochloride (NTX) is a white powder soluble in water (up to 100 mg/mL) with a peak of absorbance at 281 nm (Fig. S1, Supporting information). The UV/Vis quantification method was validated regarding linearity, accuracy and precision in water, SLF and 0.9% NaCl medium. NTX is a weak acid that at 32 °C (~ocular surface temperature) has a pKa of 8.20 due to the dissociation of the proton on aliphatic nitrogen and a pKa of 9.63 associated to the dissociation of the phenolic proton [38]. Thus, it is expected that NTX can establish ionic interactions through its positively charged aliphatic nitrogen with the negatively charged AAc. Also, hydrogen bonding may occur between AAc and the hydroxyl and carbonyl groups of NTX. It is known that in the pharmacological receptor (MOR) hydrophobic interactions contribute to stabilize ligands binding [28], and hence BzMA was chosen as an additional functional monomer (Fig. 1).


NTX easily dissolved in the monomers mixture up to 10 mM. Higher concentrations were not tested since that amount of NTX should already provide therapeutic amounts to hydrogel pieces of dimensions in the range typical of CLs. After polymerization, hydrogel sheets were immersed in boiling water (500 mL) and the amount of NTX removed was quantified spectrophotometrically at 281 nm. Boiling is typically used to clean soft CLs after fabrication, and thus both imprinted and non-imprinted hydrogels underwent the same cleaning process. Imprinted hydrogel sheets were polymerized in the presence of 10 mg of NTX (~3.3 mg/g) and nearly the whole amount of template drug was removed during boiling (Fig. 2). Differences in the UV–Vis spectra of washing medium of imprinted and non-imprinted discs were clearly observed despite the large volume of water used for boiling. Interference of residual monomers in the peak absorbance of the drug was minor (Fig. S2, Supporting information). Subsequently, the discs were immersed in SLF pH 7.8 to complete NTX removal before loading assay (Fig. 2). Salts in the SLF medium participated in ionic competition for AAc monomers and thus triggered the release of residual NTX from the polymer network. All hydrogels were prepared in triplicate, and reproducible behavior was observed.


3.2. Hydrogel characterization

Swelling degrees in water and SLF of both imprinted and non-imprinted discs were in the 46–57% range (Fig. S3, Supporting information), which is typical of HEMA hydrogels. Thus, functionalization with small proportions of AAc and BzMA did not alter water uptake. Regarding light transparency, all swollen discs showed transmittance above 85% in the visible range. A decrease in transmittance below 300 nm was recorded for NTX-loaded hydrogels (Fig. 3). This means that NTX loading is not expected to disturb a clear vision but even could protect the eye against UV radiation.


3.3. Naltrexone loading

After the drug removal step (applied to both imprinted and non-imprinted hydrogels), dried discs were immersed in a NTX aq. solution (0.3 mg/mL) to quantify the drug loading ability (Fig. 4a). Non-imprinted hydrogels without functional monomers or with only BzMA (A1 and B1) did not show any affinity for NTX. Differently, non-imprinted hydrogels with AAc could load 8.06 ± 0.27 m/g and 7.28 ± 0.41 mg/g for C1 and D1 (codes as in Table 1) respectively. The FMF values indicated that the functional monomer AAc increases the amount of NTX loaded (CFMF = 41.18) while BzMA has no influence in NTX uptake (BFMF = 0.26). The combination of AAc and BzMA led to a FMF similar to that of hydrogels with AAc (DFMF = 38.14) confirming the minor role of BzMA. Besides, imprinted hydrogels were able to increase the amount of NTX loaded (up to 10.41 ± 0.40 mg/g and 10.83 ± 0.38 mg/g for C2 and D2 respectively), with KN/W of 34.4 for C2 and 35.9 for D2. Calculated imprinting factors (CIF = 1.31 and DIF = 1.46) indicated that hydrogels synthesized with functional monomers in presence of the drug can uptake more amount of drug due to specific cavities formation inside the network. Interestingly D2 hydrogels bearing both functional monomers, although loaded NTX in a similar amount as C2 hydrogels, had a few larger imprinting factor, which means that BzMA when correctly oriented in the imprinted cavity plays a role in the loading, increasing drug affinity for the binding regions.


From a manufacturing point of view, drug loading may involve an additional, time-consuming step in the processing of drug-eluting CLs. To minimize the impact of this step, we then evaluated the feasibility of carrying out both drug loading and final sterilization in one step. First, we verified that NTX aqueous solution can withstand autoclaving by recording the UV–Vis spectrum and the drug concentration before and after sterilization (Fig. S1, Supporting information) and no significant differences were observed. Hydrogels that underwent autoclaving in the NTX solution showed similar loading (Fig. 4b) than those loaded by soaking at room temperature (Fig. 4a) although a slight increase in the amounts loaded was recorded. For comparative purposes the hydrogels autoclaved in the NTX solution were allowed to stabilize at room temperature for 24 h (i.e., under the same conditions as the non-autoclaved hydrogels). After this time, the amount of NTX loaded by autoclaved hydrogels was 8.46 ± 0.72 mg/g for C1, 8.96 ± 1.1 mg/g for D1, 11.03 ± 0.76 mg/g for C2 and 9.25 ± 0.12 mg/g for D2.


3.4. Effect of autoclaving on NTX-loaded discs

Batches of imprinted and non-imprinted discs were loaded at room temperature (as explained above) and then transferred to vials containing fresh NTX storage solution (0.3 mg/mL, 3 mL) and autoclaved (121 °C, 20 min). The purpose of this step was to elucidate whether a subsequent sterilization process may alter the amounts loaded, either by promoting further uptake or by triggering discharge. After autoclaving, samples were stored at room temperature during 2 days. Non-imprinted BzMA/AAc discs (D1) showed a minor decrease in the amount of NTX loaded after autoclaving, but it was reloaded during storage (Fig. S4a, Supporting information). Imprinted discs prepared with BzMA/AAc (D2) were able to uptake a little more amount of NTX (1.71 mg/g; Fig. S4b, Supporting information) in addition to that previously loaded. Overall, the discs retained the initial amount of NTX with minor changes during sterilization and storage.


3.5. Naltrexone release

The lack of standardized methods to evaluate drug release from ocular solid formulations, and particularly CLs, makes comparison among published data difficult. In this regard, two main setups have been identified as the most suitable ones in terms of comparison of the ability of the CLs to provide sustained release and to predict in vivo behavior. These two setups consist in either agitated sink conditions or microfluidic flow devices [11,29,30,39]. For comparison purposes, release from NTX-loaded discs was investigated using both setups. Discs that had been loaded with NTX at room temperature were rinsed with water and the excess of solution on their surface carefully wiped. Infinite sink conditions were achieved using 3 mL of 0.9% NaCl under magnetic stirring (Fig. 5). NTX was sustainably released for 24–48 h; the discs providing almost 100% drug released without showing plateaus of pseudo-equilibria [29]. Discs functionalized with AAc released higher amounts of drug to the medium than those contained in experimental eye drops (10−5 M) [6] already from the first half an hour. Imprinted discs C2 and D2 released 12.11 ± 0.33 and 13.16 ± 0.11 mg/g, respectively, and non-imprinted discs C1 and D1 released 9.90 ± 0.14 and 7.17 ± 0.05 mg/g, at day 8, in good agreement with the total amounts loaded. The well agitated conditions (200 rpm) prevented that a boundary layer was formed between the disc and the surrounding solvent, which could lead to false slow release. Thus, even under agitated sink conditions, which are prone to trigger a rapid discharge of the CLs, the NTX-loaded functionalized discs could cover one-whole day treatment.


3.6. HET-CAM and cytocompatibility tests

Potential ocular irritation effects of NTX-loaded discs and NTX were evaluated on the chorioallantoic membrane (CAM) of fertilized hen eggs. Neither the loaded discs nor NTX aqueous solution (300 μL, 0.3 mg/mL) directly placed on the CAM induced haemorrhage, lysis or coagulation, and behaved as the negative control (0.9% NaCl) (Fig. 7), which is in good agreement with the good ocular tolerance reported for NTX eye drops [2]. Cytocompatibility tests with hMSCs confirmed the high biocompatibility of hydrogels bearing AAc and BzMA after 72 h direct contact (Fig. S6, Supporting information); all hydrogels were as cytocompatible as the D2 group.



3.7. Bovine corneal permeability

Bovine corneal permeability test was carried out by monitoring the amount of NTX that diffused from the CLs (C2 discs loaded with 835.97 ± 33.02 μg, and D2 discs loaded with 866.92 ± 38.21 μg) towards the cornea and the receptor medium mimicking the aqueous humour. As a control, a concentrated solution of NTX (350 μg/mL, 2 mL) in 0.9% NaCl was used; the total amount of drug supplied to the donor compartment (700 μg) being slightly lower to the total amount of drug contained in the CLs.


After 6-h test, NTX concentration in the 0.9% NaCl solution (1 mL) of the donor chamber was 293.82 μg/mL for C2 and 295.93 μg/mL for D2 discs. In the case of the control, the corneas were exposed to a high NTX concentration since the very first minute; after 6 h test the NTX decreased to 306.56 μg/mL. Nevertheless, the amount of NTX in the receptor chamber was quite low and only quantifiable after 3 h (Fig. S7, Supporting information). This means that NTX mainly accumulated into the cornea and only a very small portion of the drug molecules (<1%) reached the receptor compartment that mimicked the aqueous humour.


Amounts accumulated in cornea were 41.23 ± 5.77 μg/cm2 for C2 and 37.66 ± 6.63 μg/cm2 for D2 discs (Fig. 8). These values were not statistically different from those recorded for the NTX solution (32.37 ± 3.91 μg/cm2) in spite that the hydrogel discs sustainedly released the drug.




4. Conclusion

Incorporation of AAc to HEMA network increases affinity for NTX through weak interactions with the aliphatic nitrogen, hydroxyl and carbonyl groups of NTX. The presence of the drug during polymerization facilitates monomers arrangement creating specific cavities that contribute to increase even more NTX affinity. Although OGFr includes hydrophobic interaction with its ligands, BzMA only plays a minor role in the loading of imprinted networks. Swelling degree, oxygen permeability and light transmission of functionalized hydrogels are in the common range for CLs. Besides, no potential ocular irritation is observed on the CAM neither cytotoxicity in hMSCs monolayer. Relevantly from the fabrication point of view, loading and sterilization can take place either simultaneously or in separate steps with minor changes in total amount loaded. Imprinted hydrogels are able to control NTX release for at least 2 days in well-agitated bulk medium and for a more prolonged period in physiological-mimicking dynamic conditions, maintaining therapeutic concentrations in the lachrymal fluid until day 3. Despite the sustained release, the NTX-loaded hydrogels allow relevant amounts of drug accumulate in the cornea in the first 6 h of application. Thus, hydrogels containing AAc as functional monomer may be suitable for preparing NTX-eluting CLs containing therapeutic amounts of drug. The effects of a small change in thickness and curvature of CLs on NTX release rate are expected to be minor but would require further investigation.

miércoles, 14 de junio de 2023

Low-dose Naltrexone - ALZHEIMER DRUG DISCOVERY FOUNDATION

Evidence Summary

Low dose naltrexone may reduce chronic pathological inflammation and hyperalgesia. It has high tolerability, but the selection of the optimal therapeutic dose may vary from person to person.

Neuroprotective Benefit: Low dose naltrexone may mitigate neuroinflammation, restore the excitatory-inhibitory balance, and promote neuronal survival. But potential efficacy may depend on the degree of endogenous opioid system dysfunction.

Aging and related health concerns: Low dose naltrexone may retrain the immune system in a manner which mitigates inflammatory damage and pain, as well as potentiate anti-tumor responses.

Safety: Tolerability is rated comparable to placebo. Reported side effects are mild and include vivid dreams, headache, and nausea. It may induce symptoms of withdrawal if taken with opioids.


Naltrexone is a synthetic orally available competitive non-selective opioid receptor antagonist. Compared to the opioid receptor antagonist, naloxone, which is used to rapidly reverse an opioid overdose, naltrexone has a longer half-life and greater oral bioavailability [2]. Naltrexone is approved for opioid use disorder to prevent someone with an opioid addiction from taking opioids by preventing the opioids from inducing the effects which make them enjoyable, and thus addictive. For this indication, naltrexone is prescribed at doses of 50 or 100 mg. However, at low doses (<5 mg), typically 1 to 4.5 mg, naltrexone has been shown to exhibit a vastly different, and in some cases, opposite, therapeutic profile. At low doses, naltrexone appears to exert anti-inflammatory and analgesic properties. This stems from the highly context-dependent nature of opioid signaling.


The endogenous opioid system primarily involves the opioid peptides, endorphins, enkephalins, and dynorphins, which interact with opioid receptors at varying affinities [3]. The major opioid receptors are mu, delta, and kappa, but there are several other related receptors that are part of the opioid receptor superfamily which can interact with this system under certain conditions. These are G-protein coupled receptors (GPCRs), which couple to various downstream signaling pathways. The expression of these receptors is dynamic and highly localized. Consequently, the downstream effects of the opioid system are highly context dependent, relative to the concentration of opioid peptides present, the composition, concentration, and localization of the receptors, the signaling effectors coupled to the receptors, as well as the presence of other factors that interact with these signaling pathways. These same factors apply to exogenous modulators of the opioid system (i.e. agonists and antagonists), including naltrexone.


Naltrexone primarily acts as an antagonist at the mu and delta receptors, but can also impact kappa receptors, to a lesser degree, and thus is classified as a non-selective antagonist. Doses that fully block these receptors can lead to receptor desensitization and associated compensatory changes, while doses which only partially block the receptor can lead to a different set of compensatory changes [3; 4].


Additionally, naltrexone is a mixture of several stereoisomers, which have differential activity toward opioid and non-opioid receptors, thus the blend of isomers in a given preparation can influence its activity [2; 3]. For example, levo-naltrexone has activity toward opioid receptors, while dextro- naltrexone engages with toll-like receptors, but not classic opioid receptors.


Pilot studies suggest that the downstream signaling and/or compensatory changes following administration of low dose naltrexone may be beneficial in conditions that involve chronic pain stemming from hyperalgesia, and maladaptive inflammation [2]. Due to the highly context-dependent nature of the opioid system, the response to low dose naltrexone would be expected to be variable, which is borne out by the pilot studies. Therefore, a highly individualized approach may be needed for the therapeutic use of low dose naltrexone.


Neuroprotective Benefit: Low dose naltrexone may mitigate neuroinflammation, restore the excitatory-inhibitory balance, and promote neuronal survival. But potential efficacy may depend on the degree of endogenous opioid system dysfunction.


Types of evidence:


4 clinical trials for high dose naltrexone/naloxone in AD


1 case series of low dose naltrexone in epilepsy


2 prescription database studies


Numerous laboratory studies




Human research to suggest prevention of dementia, prevention of decline, or improved cognitive function:


The opioid system plays important roles in learning and memory, and dysregulation of the opioid system can impair cognitive function [5; 6]. Heavy use of opioid agonists, including analgesics like morphine and drugs of abuse like heroin, as well as the use of high doses of opioid antagonists, such as naloxone and naltrexone, can result in cognitive impairment [7; 8]. These impacts to cognition can be reversible upon restoration of opioid system dynamics.


There is a body of evidence indicating that the endogenous opioid system is dysregulated in the context of Alzheimer’s disease (AD). Cerebrospinal fluid (CSF) levels of β-endorphin were found to be reduced, while levels of enkephalins and dynorphin A were found to elevated [9]. Elevated dynorphin levels have been implicated in cognitive aging [10]. The promoters of the genes encoding the mu (OPRM1), delta (OPRD1), kappa (OPRK1), and nociceptin (OPRL1) opioid receptors were found to be hypermethylated in peripheral blood cells from AD patients [11]. However, the impact of changes to circulating levels is difficult to interpret due to the highly local nature of opioid signaling. The activity of endogenous opioid peptides depends on the receptor expression, which is highly variable across cell types, thus the overall impact depends on local changes to receptor dynamics [3].


Changes to receptor expression in particular brain regions have been detected in the AD brain, including increases in the kappa receptor in limbic regions, and decreases in the mu receptor in the hippocampus [9]. Radioligand studies have indicated a reduction in global opioid receptor avidity in multiple brain regions [12]. There was a strong sex difference in the thalamus, such that the loss of receptor avidity between AD and controls was greater in women than in men [13]. This metric reflects a decrease in the number of unoccupied opioid receptors, which may indicate high receptor occupancy due to an elevation in circulating levels of opioid peptides relative to the density of receptors. This may underlie the differential responses to pain, exogenous opioids, and opioid antagonists seen in AD models and patients [8; 14].


In early trials, AD patients were found to be more sensitive to effects of opioid antagonists, naloxone and naltrexone [8; 15]. The opioid system interacts with the neuroendocrine system in mediating stress responses [5]. Chronic stress and a lack of stress resiliency are associated with AD risk. The connection between the opioid and neuroendocrine systems appears to be disrupted in the context of AD, such that the acute administration of naltrexone in AD patients fails to induce the plasma cortisol response that readily occurs in control populations [16]. The key question stemming from these findings is whether the dysregulation of the opioid system is a causal or compensatory factor in the pathophysiology of AD [5].


The opioid system is involved in the regulation of several neurotransmitter systems, impacts neurogenesis, and affects the production of neurotrophic growth factors, namely BDNF [9]. Opioid receptor activity impacts the endolysosomal system, which underlies its association with the amyloidogenic processing of APP and neuronal iron trafficking [17].


Chronic opioid drug use leads to structural and functional changes to the opioid system in the brain [9]. Evidence of AD-like pathology, namely increased levels of hyperphosphorylated tau has been detected in the brains of chronic opioid users [18; 19]. Some studies suggest levels of Aβ42 may also be increased, but this finding has been inconsistent. Endogenous opioids upregulate BDNF via the mu and delta receptors [20]. A reduction in circulating levels of BDNF is typically seen in the context of cognitive impairment, but this association is lost in opioid addicts, where elevated levels are tied to dependency, but not cognition [21]. This may be related to the tendency of opioids to mediate local/regional effects. Within the ventral tegmental area (VTA), a brain region involved in reward, BDNF is down regulated with chronic opioid use, suggestive of an increased risk for neurodegeneration, and that peripheral BDNF may be a poor indicator of CNS levels in this population [22].


Despite these findings, the association between opioid use and risk for AD is tenuous. Mortality rates for abusers of illicit drugs are higher than the general population, which may impact incidence of aging- related diseases. High exposure to prescription opioids was associated with elevated phosphorylated


tau, but not elevated Aβ42 [19]. One study found that heavy prescription opioid use (≥91 total standardized doses) was associated with a modest increase in AD risk (Hazard Ratio [HR] 1.29, 95% Confidence Interval [CI] 1.02 to 1.62), compared to <10 doses, but heavier opioid use was not associated with faster rates of cognitive decline [23]. Lower levels of opioid use (<91 total standardized doses) were not significantly associated with elevated AD risk. A separate study found that chronic prescription use of opioids (>90 total standardized doses) was not significantly associated with increased risk for AD (adjusted Odds Ratio [OR] 1.02, 95% CI 0.98 to 1.07). However, prescription use of opioids occurs in the context of injury and pain disorders, which typically involves inflammation, that is itself a feature associated with AD risk. Thus, the immune regulatory and anti-inflammatory effects of opioids may actually serve to mitigate risk in this population. Meanwhile, the changes leading to opioid tolerance are associated with increased levels of neuroinflammation [9], which may then elevate risk. Consequently, the impact of exogenous opioid use depends on a variety of individual factors, such as the baseline state of the endogenous opioid system and the duration/intensity of use.


Altogether, the data suggest that the impact of opioids on dementia risk is highly complex. The alterations to the endogenous opioid system are most likely compensatory. However, at least some of these changes appear to contribute to cognitive decline or disease progression. Thus, there may be utility in preventing the detrimental compensatory changes to the opioid system. Since endogenous opioids appear to have a variety of anti-inflammatory and neuroprotective activities, individuals with low opioid tone may be at elevated risk, and interventions which may help normalize opioid tone, such as low dose naltrexone, may be beneficial in mitigating risk.




Human research to suggest benefits to patients with dementia:


The dysregulation of the opioid system may contribute to AD pathophysiology and cognitive impairment. However, due to the complex and region-specific nature of the dysregulation, it is unclear whether a broad-acting or non-selective opioid receptor modulator would offer significant therapeutic utility [5]. A more viable therapeutic option may be a combination of selective modulators. Naltrexone acts primarily at the mu and delta receptors [2]. Since, at low doses, it paradoxically acts more like a weak agonist than an antagonist, low dose naltrexone may be useful as part of an opioid system normalizing therapeutic regimen. However, caution is warranted in the selection of a potentially therapeutic dose in this population. Low dose naltrexone refers to doses less than 5 mg/day, with 4.5 mg as the most widely tested dosing regimen. Studies using naloxone and naltrexone suggest that AD patients are more sensitive to these opioid antagonists [8], such that lower doses may be needed.


Though there is also evidence from case reports that naltrexone can inhibit drug/alcohol seeking behavior in individuals with dementia and addiction disorders, suggesting that naltrexone may still be able to reliably influence the opioid system in at least a subset of dementia patients [24]. Careful dose titration studies would be necessary. It is also possible, as suggested by the altered cortisol response to high dose naltrexone [16], that in some AD patients, the extent of dysregulation to the opioid system may be so severe that the normalizing effects seen with low dose naltrexone in other populations may not be possible at any dose.




However, studies in other indications suggest that the mechanism of benefit for low dose naltrexone may extend beyond modulation of the endogenous opioid system, and extend into the modulation of receptor systems that are part of the broader opioid receptor superfamily [2; 3]. The best characterized of these is the modulation of the immune system via toll-like receptors (TLRs). Dysfunction of TLRs has been implicated in AD, cognitive aging, and a variety of ‘inflammaging’-associated chronic age-related diseases. Consequently, low dose naltrexone could potentially benefit AD patients by reducing deleterious neuroinflammation. Due to the dysfunction of TLR responses in AD and with aging, it is unclear whether low dose naltrexone could mitigate inflammation to a similar degree as has been seen in other patient populations. Overall, the potential benefit of low dose naltrexone likely declines with disease severity, and may be most useful during the early stages of the disease when low dose naltrexone may be most able to normalize the system.


 



Alzheimer’s disease: (COGNITION) HIGH DOSE NALTREXONE – NO BENEFIT


In the 1980s, several clinical trials were conducted testing naloxone or naltrexone at doses typically used for drug addiction [8; 15; 25; 26]. These patients were clinically diagnosed with dementia of probable


Alzheimer’s type. Most studies found no improvement on cognitive measures. It was noted in the naloxone studies, which is administered intravenously, that the responses were different from what is typically seen in young healthy adults. The dementia patients experienced sedation at the highest doses and agitation at the lower tested doses, and any purported cognitive enhancing effects seen may have been related to increased stimulation/agitation in the patients, rather than a true effect on cognition [8; 15]. The lowest tested dose for naltrexone was 5 mg, thus these studies do not provide good insight into the potential effects of low dose naltrexone in this population [25].




The efficacy of naltrexone on patients in the early stages of cognitive decline, such as those with mild cognitive impairment, has not been established. At early stages, the level of dysfunction within the endogenous opioid system may be as amenable to therapeutic intervention as other conditions with altered opioid function, such as chronic pain and mood disorders. Normalization of opioid tone could offer a wide range of therapeutic effects, including impacts to synaptic function, excitatory-inhibitory neurotransmitter balance, neurogenesis, cellular stress resiliency, and immune modulation [9].


Biomarker-based studies assessing these outcomes would be needed to determine the potential therapeutic utility of low dose naltrexone in this population.





Alzheimer’s disease (AGITATION): POTENTIAL BENEFIT FOR PAIN MANAGEMENT (theoretical) Historically, antipsychotic medications have been widely used to treat agitation and behavioral symptoms of AD. However, due to an increased understanding of the dangers of these drugs, prescribing practices have shifted. In association with the decrease in antipsychotic use, there has been a marked increase in the use of opioids in this population [27; 28; 29]. Part of this stems from efforts toward better pain management in this population, and a recognition that the presentation of agitation could be a reflection of uncontrolled pain. They may also be used for this indication due to their sedative properties. It is unclear whether the chronic use of opioid medication could be detrimental in this population, as it has not been well studied. Evidence indicating dysfunction of the endogenous opioid system suggests that the therapeutic profile is likely to be altered in this patient population. Since several of the brain adaptations to heavy opioid use are consistent with the types of changes and pathophysiology seen in the context of AD [18; 19], it is possible that the persistent use of opioids for agitation could exacerbate pathology and accelerate functional decline.


In non-demented populations, chronic use of low dose naltrexone has been shown to have analgesic, but not sedative properties, presumably via modulation of the endogenous opioid system [2]. If low dose naltrexone had similar analgesic properties in AD patients, then it could potentially be a safer alternative for pain management. Due to dysregulation of the opioid system in AD patients, the translatability of the effects seen in patients without dementia is unclear. Identifying the optimal dose would be critical, as prior studies using moderate dose naloxone/naltrexone found evidence for increased agitation [8]. This could be due to a potentiation of pain due to the blockage of opioid receptors, an effect on the regulation of wakefulness, or a combination of factors. More studies are needed to determine if a low or ultra-low dose of naltrexone can be safe and effective in this population.




Mechanisms of action for neuroprotection identified from laboratory and clinical research:


The opioid system intersects with a variety of other systems that are impacted or implicated in dementia. Some key areas include the regulation of various neurotransmitter systems, the regulation of cellular metabolism via PI3K/Akt and mTOR, and the modulation of the immune system [5; 9; 30]. While it remains to be established whether low dose naltrexone is neuroprotective, its off-label use in a variety of indications offers suggestive evidence that it has the potential to modulate these parameters in a clinically beneficial manner.





Amyotrophic lateral sclerosis: UNCLEAR BENEFIT


In a cross-sectional questionnaire of off-label medication use in patients with ALS (n=41) in Norway, low- dose naltrexone was identified as the most commonly used off-label medication (n=8, 19.5%) [31].


Relative to the total study population, those using low-dose naltrexone reported a better physical components score on the self-reported RANDS-12 questionnaire.





Epilepsy: POTENTIAL BENEFIT TO REDUCING SEIZURES


A case series of five children (ages 6-15) in Egypt with intractable epilepsy (5-10 seizures/day) found that low-dose naltrexone (1-5 mg/day) reduced seizure frequency/ epileptiform activity in these children, with two of the children remaining seizure-free for at least three months after starting low dose naltrexone [32].


A controlled study utilizing the Norwegian Prescription Database (n=11,247) found that there was a dose-response relationship regarding exposure to low dose naltrexone and use of antiepileptics, antipsychotics, and antidepressants [33]. Overall, the number of antiepileptic users decreased by 3.1% points, (95% CI 1.6% to 4.6%, p < 0.001). This was primarily driven by regular use of low dose naltrexone, defined as having filled at least four prescriptions for it. In this group, the number of users decreased by 1.7% points (from 807 to 722). Amongst those using antiepileptic medication, use of low dose naltrexone was not significantly associated with a reduction in daily dose.


The impact on seizures may be related to its effects on endogenous opioid signaling and the modulation of the immune system. Endogenous opioids show anticonvulsant properties. Low dose naltrexone may boost levels of endogenous opioids as part of a compensatory response to an acute/partial receptor blockade. Consistent with this proposed mechanism, ultra-low dose naltrexone has been shown to potentiate the anticonvulsant properties of the opioid agonist, morphine [34].





Psychiatric disorders: POTENTIAL/UNCLEAR BENEFIT


In a register-based study involving the Norwegian Prescription Database (n=11,247), persistent use of low dose naltrexone was associated with a reduction in the use of antidepressants and antipsychotics [33].


Persistent use of low dose naltrexone was associated with a reduction in the use of antipsychotic medications. Within the two-year analysis period, there was as 25% increase in the use of antipsychotics for those with the lowest exposure to low dose naltrexone, but a 17% decrease in their use in those with the most consistent exposure (≥ 4 low dose naltrexone prescriptions). Additionally, amongst users of antipsychotics, the defined daily dose (DDD) of antipsychotics was reduced by 11% in individuals with the highest exposure to low dose naltrexone, relative to those with the lowest exposure. There was also a dose-response relationship in the use of antidepressants, with a 2% reduction in those with low naltrexone exposure, and a 21% reduction in those with more consistent exposure, but no effect on the DDD amongst users of antidepressants. Low dose naltrexone use did not reduce the use of anxiolytics or hypnotics. Since this study was not tied to any particular condition, it is unclear whether these effects are related to changes in the use of other medications, such as opioids, that influence psychiatric parameters, or if they reflect an effect on brain physiology.


Some studies testing low dose naltrexone in conditions where depression/mood disorders are common comorbidities have been variable with respect to whether low dose naltrexone was found to impact mood [35; 36].


APOE4 interactions: Not established


Aging and related health concerns: Low dose naltrexone may retrain the immune system in a manner which mitigates inflammatory damage and pain, as well as potentiate anti-tumor responses.




Types of evidence:


2 systematic reviews of studies testing naltrexone in cancer


2 systematic reviews of studies testing low dose naltrexone in chronic pain disorders


1 systematic review of RCTs in inflammatory bowel disease


3 clinical trials in fibromyalgia


1 clinical trial in diabetic neuropathy


3 prescription database studies


Numerous laboratory studies






Chronic pain disorders: POTENTIAL BENEFIT


The off-label use of low dose naltrexone has primarily occurred in the context of chronic pain disorders. Since most of the studies have been small proof-of-principle type, the therapeutic benefit of low dose naltrexone has not yet been conclusively demonstrated in any population. The studies illustrate a high degree of variability in response, in terms of the degree of response (none, partial, full), and the length of time needed to achieve a response [36; 37]. In most cases, several weeks of treatment was needed for a response to occur, suggesting that the effects were related to adaptive changes and remodeling within the endogenous opioid system network.


There are two major, non-mutually exclusive, hypotheses as to the mechanism by which low dose naltrexone exerts its therapeutic effects in pain disorders [2]. One mechanism involves the rebalancing of the endogenous opioid system. Partial blockade of the opioid receptors, as occurs with low doses of naltrexone, can lead to a compensatory increase in the production of endogenous opioids. This process may also lead to changes to the expression and localization of the opioid receptors. Together these changes may increase the endogenous opioid tone, leading to a reduction in hyperalgesia/sensitivity. This may impact mechanisms involved in central sensitization implicated in neurogenic pain, such as glial activation and neuronal hyperexcitability [36]. It may also impact inflammation-associated pain, as endogenous opioids are known to regulate immune system responses. Alternatively, or additionally, low dose naltrexone may modulate the immune system and mechanisms of inflammatory pain via other receptors in the same superfamily of opioid receptors, such as TLRs. Various studies have found that low dose naltrexone can impact TLR4, though there is also evidence that it may interact with a variety of TLRs, including TLR2 and TLR9, depending on the tissue environment and cell type [2; 3; 38]. The modulation of mTOR activity has been implicated as a key mechanism by which low dose naltrexone influences the metabolic/activation state of microglia and macrophages [39].


A better understanding of the mechanism by which low dose naltrexone exerts its therapeutic effects may allow for the identification of the patients who are most likely to benefit from this treatment. In the studies conducted thus far, there were responders and non-responders, but reliable indicators of prospective responders are currently lacking [36; 37]. One small study (n=78) found that patients with neuropathic pain showed more benefit than those with inflammatory pain, but the groups were not balanced at baseline [40]. Disorders of chronic widespread pain, which may be indicative of systemic dysfunction in endogenous analgesic pathways, are more likely to benefit than conditions with acute localized pain [36]. Dosing is another potential source of variability. Opioid signaling is highly context dependent, and the therapeutic benefits of low dose naltrexone appear to depend, at least in part, on the degree of receptor inhibition. There seems to be a critical sweet spot of partial inhibition. Due to differences in the state of the endogenous opioid system across both patient groups and individual patients, the optimal dosage may vary from condition to condition or from patient to patient.





Fibromyalgia: POTENTIAL BENEFIT IN A SUBSET


Fibromyalgia is a chronic pain disorder involving musculoskeletal pain, fatigue, sleep disturbances, and mood disorders, which primarily affects women. The etiology is unclear, but patients with fibromyalgia have been shown to have low opioid tone, which may result in a hyperalgesia phenotype [35]. Several pilot studies have shown benefit for low dose naltrexone in patients with fibromyalgia [36].


In a single-blind crossover study including 10 women with moderate fibromyalgia, treatment with low dose naltrexone (4.5 mg/day) for eight weeks reduced patient-reported pain symptoms by 32.5%, relative to the placebo period [41]. Six out of the eight participants were classified as responders, which was defined as greater than 30% pain reduction relative to placebo. Drug responsiveness was positively correlated with erythrocyte sedimentation rate, a measure of inflammation (0.91, P < 0.0005).


Significant effects were seen on pain, stress, and fatigue, but not on sleep. Mechanical and thermal pain thresholds were also increased, while cold pain thresholds were unaffected. Similar results were seen in a 22-week randomized, double-blind, placebo-controlled crossover trial including 31 women with fibromyalgia [42]. Self-reported pain was significantly reduced with low dose naltrexone (4.5 mg) relative to placebo (28.8 ± 9.3% reduction vs 18.0 ± 10.8% reduction). General satisfaction (11.1% versus 3.2%) and mood (10.7% versus 2.1%) were also significantly improved, while sleep and fatigue were not significantly affected. Nine patients (32%) treated with low dose naltrexone, relative to three (11%) on placebo met all criteria for a positive response. The impact of low dose naltrexone on systemic plasma biomarkers of inflammation was assessed in a single-blind crossover trial including eight women with fibromyalgia [43]. After correcting for multiple comparisons, levels of the cytokines associated with inflammatory pain, TNF-α, IL-1β, IL-2, IL-6, IL-15, and IL-17, were found to be reduced following eight weeks of treatment. Patient-level data was provided only for TNF-α, which indicated that the response was variable, with 4/8 showing a clear downward trend, 2/8 showing no major change, and 2/8 showing an upward trend. For the majority of these inflammatory markers, at least six weeks of treatment were needed to see a meaningful reduction.


Altogether these studies suggest that low dose naltrexone may benefit a subset of patients with fibromyalgia. Fibromyalgia may be an umbrella term for a constellation of similar pain disorders with distinct etiologies, and the pilot studies suggest that not all patients with a diagnosis of fibromyalgia are likely to benefit equally. The inflammatory profile may influence responsiveness, but a predictive biomarker panel for responsiveness has yet to be validated. A set of Phase 3 RCTs (the INNOVA study and the FINAL study) are currently underway in Spain and Denmark to validate the pilot study findings regarding the use of low dose naltrexone (4.5 mg/day) in fibromyalgia [44].





Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: POTENTIAL BENEFIT


ME/CFS is characterized by persistent fatigue of unknown etiology that involves neurological and immunological dysfunction. A retrospective medical records-based study including 218 patients with ME/CFS treated with low dose naltrexone (3-4.5 mg/day) found that 73.9% of the patients reported a beneficial response [45]. The most common responses were improved vigilance/alertness (n=112; 51.4%), improved physical performance (n=52; 23.9%), improved cognition (n=46; 21.1%), pain relief (n=36; 16.5%), and less fever (n=33; 15.1%).


TRPM3 is a nociceptive ion channel that is regulated by opioid receptor activity. TRPM3 activity was found to be altered in Natural Killer (NK) cells (CD3-/CD56+) from patients with ME/CFS (n=9) relative to healthy controls (n=9) [46]. In response to treatment with low dose naltrexone (average dose 4.06 ±


0.68 mg/day), TRPM3 channel activity in NK cells was normalized. This study did not correlate these immune parameters with functional/symptomatic outcomes, so it is not clear whether this is a mechanism of drug efficacy in this population.





Arthritis: POTENTIAL BENEFIT IN A SUBSET


A controlled, quasi-experimental study utilizing the Norwegian Prescription Database (n=360) examined the impact on prescriptions of low dose naltrexone on the use of traditional disease-modifying and analgesic medications in patients with rheumatoid and seropositive arthritis [47]. The study found that patients who had received four or more prescriptions of low dose naltrexone had a 13% relative reduction in the cumulative defined daily dose (DDD) of all examined medications (-73.3 DDD per patient; 95% CI -120.2 to -26.4, p = 0.003). There were significant reductions in the DDD of NSAIDs, and opioids, as well as significant reductions in the number of patients using disease-modifying drugs (DMARDs), TNF-α antagonists, and opioids. Acute use of low dose naltrexone (≤ 2 prescriptions) was not associated with medication reductions. Although the reduction in use of opioids, presumably for pain, may be due to the contraindication between naltrexone and opioids, the overall trend suggests that consistent use of low-dose naltrexone may improve arthritis symptoms, resulting in a reduction in other symptom-modifying medications.





Inflammatory bowel disease: POTENTIAL BENEFIT IN A SUBSET


Low dose naltrexone has been tested in patients with inflammatory bowel diseases (IBD), including Crohn’s disease, and ulcerative colitis. A quasi-experimental study of the Norwegian Prescription Database found 582 patients with IBD who had at least one prescription for low dose naltrexone, 256 of which were classified as persistent users, having filled at least four prescriptions [1]. In this population, use of intestinal anti-inflammatory agents was reduced by 17%, use of intestinal corticosteroids was reduced by 32%, while use of other immunosuppressants was reduced by 29%, compared to the two years prior to starting naltrexone. There were no significant dose reductions amongst those who maintained use of these anti-inflammatory drugs. A Cochrane systematic review including two RCTs


testing low dose naltrexone in Crohn’s disease found that there was insufficient evidence regarding efficacy in this population [48]. One study assessed low dose naltrexone (4.5 mg/day) relative to placebo for 12 weeks (n=34) in adults. The difference in clinical remission between the groups was not statistically significant (30% vs 18%) (Relative risk [RR] 1.48, 95% CI 0.42 to 5.24), though there was a significant improvement in patients achieving a 70-point clinical response (83% vs 38%) (RR 2.22, 95% CI


1.14 to 4.32). The proportion of patients achieving an endoscopic response was also higher in the drug group (72% vs 25%) (RR 2.89; 95% CI 1.18 to 7.08), but no significant difference in rates of endoscopic remission. The other study included 12 pediatric patients treated for eight weeks, of which 25% of low dose naltrexone (0.1 mg/kg) users achieved clinical remission, compared to none in the placebo group. In an uncontrolled prospective study including 47 patients with refractory IBD, treatment with low dose naltrexone (4.5 mg/day) for 12 weeks resulted in temporary improvement for 48.9% (n=23) of patients, and remission for 25.5% (n=12) [49]. Of the six patients with clinical remission tested endoscopically, five were found to also exhibit endoscopic remission. Serum levels of IL-8 and TNF-α were not significantly altered by treatment in responders or non-responders. Intestinal tissue biopsies and organoids showed that ER stress, as measured by GPR78 levels, was reduced in response to naltrexone treatment.


All together these studies suggest that low dose naltrexone may benefit a subset of patients with IBD, though it is unclear how to identify individuals who might benefit. A randomized, double-blinded, placebo-controlled multicenter trial is currently underway to test the ability of low dose naltrexone (4.5 mg/day) to induce endoscopic remission within 12 weeks [50].





Cancer: POTENTIAL BENEFIT AS ADJUNCT AND FOR PAIN


Low dose naltrexone has been used in the management of cancer-associated pain. There are several case reports suggesting that low dose naltrexone may also have anti-cancer properties, and may be a useful adjunct to traditional anti-cancer therapies [4; 51]. Preclinical studies highlight the critical role of dosing in the cancer-related activity of naltrexone.


Case reports in a variety of cancer types have identified cases where the use of low dose naltrexone as a supplement to standard therapy, led to clinical remission and/or an extension of overall survival [51]. In clinical studies, low dose naltrexone reduced the toxicity and improved the tolerance of chemotherapeutics, without negatively impacting their anti-cancer activity [4]. Preclinical studies suggest that low dose naltrexone may improve the efficacy of anti-cancer therapies by potentiating the adaptive immune response, while also reducing the chronic low-grade inflammation in the tumor microenvironment that is conducive to its growth and maintenance [3; 35].


Endogenous opioids are involved in the regulation of cell growth, and depending on the context, they can potentiate or inhibit cancer cell growth. Opioid growth factor (OGF) and its receptor (OGFr) may be the major mediators of opioid effects in cancer, as the expression of this system is altered with disease progression, and has been associated with disease severity [4]. The beneficial role of low dose naltrexone in cancer appears to be dependent on a partial/intermittent blockade of the opioid receptors. A transient blockade may lead to a decline in a program promoting DNA synthesis. A rebound of endogenous opioids during the withdrawal period may prime the system for a more persistent inhibition of tumor cell proliferation [35]. The mechanisms of action have not been fully elucidated, and likely involve a wide variety of compensatory changes. High doses of naltrexone, where receptors are fully blocked promotes tumor growth in preclinical models [3; 4]. Only intermittently administered low dose naltrexone generates an anti-tumor response in these models [2].


Well-designed clinical studies are needed to determine the translatability of these findings, including the optimal dosing strategy in patients, potential biomarkers of response, and whether particular chemotherapeutics or immunotherapies work synergistically with low dose naltrexone.





Diabetes: POTENTIAL BENFIT FOR PAIN (Clinical) AND GLUCOSE TOLERANCE (Preclinical)


Low dose naltrexone (2-4 mg/day) was compared to amitriptyline (10-50 mg) in a randomized, controlled, crossover trial in 67 patients with painful diabetic neuropathy for six weeks [52]. The change in the visual analog scale (VAS) for pain from baseline between the groups was not significantly different (1.64, 95% CI −0.92 to 4.20), and the use of rescue medications was similar between groups. While the pain management was similar between the two drugs, the safety profile of naltrexone was superior, with only eight reported adverse events, compared to 52 for the tricyclic amitriptyline. This study did not assess changes to other diabetes-related parameters, but evidence from preclinical studies suggests that low dose naltrexone may exert benefits to diabetic patients beyond pain relief.


In a high-fat diet-induced model of type 2 diabetes, treatment with low dose naltrexone (1 mg/kg/day i.p.) for the last two weeks of a five-week period of high-fat diet, improved glucose tolerance and insulin sensitivity in male mice [53]. It attenuated the release of pro-inflammatory cytokines typically associated with hyperinsulinemia, by enhancing the deacetylase activity of SIRT1 and inhibiting the activation (nuclear localization) of NF-kB. In the same model and treatment regimen, low dose naltrexone also improved bone quality parameters [54; 55]. Type 2 diabetes is associated with reductions in bone hardness, mineral composition, collagen content, and size. These macro and microstructural changes result in weaker, fracture-prone bones. Treatment with low dose naltrexone improved bone hardness, mineral size, the mineral-to-matrix ratio, collagen content, and lowered the level of advance glycation end products (AGEs) in the bone. If similar effects were found to occur in humans, low dose naltrexone could potentially lead to improvements in pain, glucose tolerance, and bone strength in type 2 diabetics. Clinical studies are needed to determine the translatability of these effects, and whether they are related to the immune modulatory activity of low dose naltrexone, or involves other mechanisms.


Safety: Tolerability is rated comparable to placebo. Reported side effects are mild and include vivid dreams, headache, and nausea. It may induce symptoms of withdrawal if taken with opioids.




Types of evidence:


1 meta-analysis of RCTs testing naltrexone assessing adverse events


2 systematic reviews of studies testing low dose naltrexone in chronic pain disorders


1 systematic review of RCTs testing low dose naltrexone in inflammatory bowel disorders


Numerous laboratory studies




Naltrexone is generally considered safe, and even at the doses used for addiction disorders, 50 and 100 mg, it is associated with few side effects. Opioid antagonists, such as naltrexone, are considered much safer than opioid agonists, for which overdoses can be lethal. Common side effects with naltrexone include nausea, headache, dizziness, anxiety, and insomnia, but are generally mild (WebMD). A meta- analysis of 89 RCTs including 11,194 participants testing naltrexone (at any dose range) found that there was no significantly increased risk for severe adverse events with naltrexone relative to placebo (RR 0.84, 95% CI 0.66 to 1.06) [56]. Subgroup analysis indicated that these findings held across dosages and disease groups.


The levels of all adverse events were generally matched (3,938 in the naltrexone arm and 3,079 in the placebo arm), and events were reported as mild or moderate.


In general, the side effect profile for low dose naltrexone appears to be better than for higher dose naltrexone. The incidences of adverse events were similar relative to placebo. Tolerability was generally rated near 90% and in some cases was rated as more tolerable than the placebo [2; 43]. In pilot studies vivid dreams and sleep disturbances were rated as the most common adverse events [2]. These tended to occur shortly after starting treatment, and decrease over time. In most studies, the naltrexone was administered right before bed, which may account for the impacts to sleep. Altering the timing of administration to the morning has been suggested as an alternative for individuals experiencing sleep- related effects [49]. In chronic pain disorders, disease-related sleep disturbances were generally resistant to treatment with low dose naltrexone, which may be related to naltrexone’s impact on sleep [33; 36]. Headache and gastrointestinal effects, such as nausea, were the next most common adverse events [2; 4; 36; 37; 48; 49; 52]. These were generally rated as mild, and diminished over time.


Drug interactions: The use of opioid agonists is contraindicated with the use of naltrexone, as it could precipitate symptoms of opioid withdrawal (Drugs.com).


 


Sources and dosing:


Naltrexone is FDA-approved for opioid use disorder and alcohol use disorder. It is available as an intramuscular injection (Vivitrol®) and in the form of extended-release 50 mg tablets (ReVia®). For this indication it is typically dosed at 50 or 100 mg/day. It is available in both branded and generic formulations. Low dose naltrexone refers to doses less than 5 mg between 1 to 4.5 mg, while ultra-low doses (microdoses) refer to doses less than 0.1 mg. The most commonly tested ‘low dose’ is 4.5 mg.


There appears to be a relatively tight therapeutic dose window for low dose naltrexone, which may vary from person to person. Low dose naltrexone is not currently approved for any indication, and is only available when prescribed off-label. Additionally, preparations of low dose naltrexone need to be obtained from compounding pharmacies, because approved tablets are manufactured at 50 mg.




Research underway:


According to Clinicaltrials.gov, there are currently 14 active clinical trials testing low dose naltrexone. Low dose naltrexone is being tested for Fibromyalgia, Complex Regional Pain Syndrome, Covid-19, Vasculitis, Endometriosis, Bladder Pain, Diabetic Neuropathy, Aging (observational study), and Postural Orthostatic Tachycardia Syndrome (POTS). Additionally, there are trials in Europe testing low dose naltrexone in Fibromyalgia and in Inflammatory Bowel Disease.


AgelessRx, which provides prescriptions and supplements for longevity-related products is sponsoring a retrospective observational study assessing the effects of short, intermediate, and long-term (>5 years) use of low dose naltrexone (<20 mg/day) on general health and immune parameters, as well as markers of phenotype age based on blood biomarkers (NCT05307627).


ESTUDIO ACTUALIZADO A FECHA 24 MAYO 2022