Dementia is one of the largest public health and social care challenges facing today’s society. Alzheimer’s disease is the most common neurodegenerative disorder resulting in progressive cognitive decline, affecting more than 50 million individuals globally, and expected to triple in the coming thirty years [1]. The current symptomatic therapeutics for Alzheimer’s disease (acetylcholinesterase inhibitors and memantine) have been shown to have limited efficacy and sometimes challenging side effects [2]. A systematic review and meta-analysis on the efficacy and safety of the available symptomatic treatments for patients with mild cognitive impairment showed that the current marketed products did not substantially improve function and cognition in this patient group and are associated with risks [2].
Aiming for disease modification by targeting the amyloid cascade has been the selected strategy for many drug development programs throughout the last 25 years [3, 6]. Encouraging clinical study data on the efficacy of antibodies targeting the amyloid cascade as treatment for Alzheimer’s disease are emerging [4, 5, 6]. Aducanumab and lecanemab have already been granted approvals from the US Food and Drug Administration and donanemab is expected to be filed in 2023. Although promising, it remains to be seen how widely these antibodies will be used in clinical practice. Even considering the availability of effective antibodies, a need for additional treatment options to manage Alzheimer’s Disease remains. A new effective and safe symptomatic treatment with cognitive enhancing effects to be used alone or in combination with treatments such as the antibodies would be of great benefit to patients.
The importance of neurotrophin signaling pathways in the human brain has been well established. Pathways involving nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF) are of paramount importance for neuronal cell function, communication, and cell survival in brain areas vital for cognitive function, such as the hippocampus and basal forebrain. BDNF and NGF exert their effects through binding to their cognate tropomyosin receptor kinase (Trk) receptors; TrkA or TrkB, respectively [7, 8]. Pathological and mechanistic evidence suggests that loss of NGF signaling contributes significantly to the dysfunction of basal forebrain cholinergic neurons in Alzheimer’s disease [9, 10]. Decreased levels of BDNF have been observed in the hippocampus and in cerebrospinal fluid in disease states with cognitive decline, including Alzheimer’s disease [11, 12, 13], indicating that decreased BDNF signaling may contribute to this cognitive decline. The transplantation of stem cells or lentiviral delivery of BDNF into the brain of amyloid-transgenic mice or primates resulted in reversal of synapse loss and in improved cognition [14]. Furthermore, accumulating evidence suggests that increased BDNF signaling could improve cognition in Alzheimer’s disease [15, 16], indicating that positive modulation of NGF and BDNF signaling may be a relevant strategy to develop new treatment options for Alzheimer’s disease and other conditions where cognition is impaired.
The NeuroRestore® program, initiated by AlzeCure Pharma AB, Sweden, aims to identify small molecules with a stimulatory effect on BDNF and NGF signaling. As reported by Dahlström et al. in 2021 [17], two lead molecules were identified as effective positive allosteric modulators of Trk-receptors; ACD855 and ACD856. ACD855 is identical to the veterinary product Ponazuril, a triazine-based drug that acts to inhibit enzyme systems in protozoa and is used for the treatment of equine protozoal myeloencephalitis [18]. Both compounds demonstrated pro-cognitive effects in various preclinical in vivo models in mice [17, 19]. Cognitive enhancement was also seen in aged animals with a natural decline in memory, where a single dose of ACD856 led to an improved memory [17]. Furthermore, both ACD855 and ACD856 have also demonstrated anti-depressant like effects in the forced swim test model [20].
The clinical efficacy, safety and pharmacokinetics (PK) of ACD855 has been evaluated in various domestic animals for the use as a veterinary drug [21]. ACD855 was further investigated in general toxicity studies in rats and dogs with treatment duration up to 28 days duration and assessment of recovery for 2 weeks, and there was no indication of any mutagenic or genotoxic potential. The PK of ACD855 in various animals indicated that ACD855 was rapidly absorbed, followed by a prolonged elimination, with terminal half-life in plasma between 0.3 to 7.9 days in animals. The elimination half-life in man was predicted through allometric scaling to 4.6 days. Based on pharmacology studies in rodents and safety studies in rat and dog, the first oral dose in an initiated Phase I single ascending dose (SAD) study (EudraCT No. 2018-002320-16), in healthy volunteers was set to 1 mg. After completion of cohort 1, the elimination half-life in plasma in humans was found to be significantly longer than predicted, with an average of 68 days, Figure S1. The study was prematurely terminated after interim PK analysis of the first cohort, and long-term follow-up of the dosed subjects was performed. The plasma exposure of ACD855 declined slowly over time and all subjects but one still exhibited levels above the detection limit of 1 ng/mL at 280 days when the safety review committee decided that, in the absence of any safety concerns and the fact that the predicted AUCinf was lower than the NOAEL in the most sensitive species, follow up could be concluded.
The follow-on compound ACD856 was selected based on in vitro drug metabolism and PK properties, in addition to its demonstrated improved potency compared to ACD855. This included an improved profile with regards to metabolic characterization in hepatocytes and liver microsomes and in vivo PK in animals, predicting a shorter elimination half-life in humans.
This paper describes the start of the ACD856 clinical development program, including first drug administration in humans in an i.v. microdose study (NCT05783830) and an oral SAD study (NCT05077631). The aim was to evaluate the safety, tolerability, and pharmacokinetics following single doses of ACD856 in healthy subjects. In addition, the impact of food intake on PK properties was evaluated after oral dosing. Prior to initiating the microdose study, a limited program of toxicology studies was performed, in accordance with current guidelines and regulations. This included an evaluation of the genotoxic potential in silico, and an extended single i.v. bolus dose toxicity study in rats. Additionally, due to notable clinical signs consisting of gastrointestinal disturbance in dogs with ACD855, a seven-day oral dose range finding study in minipigs was conducted to assess the tolerability of ACD856 within this species. Prior to initiating the SAD study, ACD856 was tested in accordance with regulatory standards, including studies such as genotoxicity, general and safety pharmacology and 28-day toxicology studies in rats and minipigs. No safety findings were found at highest dose levels tested up to 28 days in both species (assessments included body weight, food consumption, ophthalmology, coagulation parameters, electrocardiogram (ECG), hematology-, clinical chemistry- and urinalysis). An investigation into potential target related adverse effect was done by reviewing publications from clinical studies of treatment approaches with similar modes of action i.e., stimulating NGF or BDNF signaling [22, 23, 24, 25, 26, 27, 28]. In these studies, changes of gut motility (loose stools and increased frequency of stools), reduced appetite and weight loss, diffuse myalgia and eosinophilia were observed as adverse effects potentially related to the mechanism of action. None of these adverse effects had been described as severe or serious adverse events.