White Paper

The Use of Robust Biomarkers in the Early Clinical Development of N-methyl-D-aspartate (NMDA) Receptor Antagonist

Biotrial has extensive early clinical development experience with a high output of over 20 CNS studies per year. The experience and capabilities of Biotrial Neuroscience (now also incorporating the know-how of the legacy CNS CRO Forenap), enables the in-depth cognitive and neurophysiological assessment of NMDA antagonists. The aim of the present document is twofold. Firstly, to characterize the pharmacological effects of non-competitive NMDA antagonists in psychiatry and pain indications and to validate the use of as a positive control in the development of these compounds with this mechanism of action. Secondly, to present Biotrial’s strategy and rationale for the integration of surrogate endpoints, biomarkers and PD models into a Phase I program, in order to establish early proof of concept of new NMDA antagonist candidates. Pharmacological Effects of Non-Competitive NMDA Antagonists: The Example of Ketamine

Glutamatergic antagonists, such as ketamine, acting at the NMDA open channel have a long history of use in neuropsychiatry – independently of their anesthetic or pain attenuating properties. The mechanism of action and pharmacological effects of ketamine have been extensively studied over the last 20 years. Ketamine was initially profiled as a reversible model of psychosis1, producing positive and negative symptoms of schizophrenia as well as cognitive impairment. As of today there are approximately one hundred publications on the symptoms, brain connectivity, imaging and cognitive function alterations induced by ketamine. The main hypothesis to explain the effect of ketamine was the reduction of the burst in the prefrontal cortex, secondarily enhancing AMPA α-(amino- 3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor) signaling. The mammalian rapamycin (mTOR) signaling pathway and its subsequent release of brain-derived neurotrophic factor (BDNF), possibly allowing the formation of new synapses, has also been reported as a potential target. Lastly, evidence shows activation of the early activity gene (Arc), which is known to down-regulate AMPA receptors. AMPA receptors are, in turn, part of oscillation-forming thalamo-cortical loops2,3, which may partially explain the increased effects on qEEG signals observed in animals4.

Psychiatry Applications

More recently, reports of long-lasting improvements of depressive symptoms with ketamine5 have led to the development of related compounds with less of the psychotomimetic effects and the assumed effect based on animal data, such as AZD6765 (), a low-trapping NMDA receptor antagonist6,7. Comparable effects should be achieved with NR2B antagonists ( and MK-0657) or with mGluR2 and mGluR5 Negative Allosteric Modulators8. A recent meta-analysis of 5 studies which assessed the effects of ketamine in Major Depressive Disorders (MDD) in 134 depressed patients9, showed that ketamine was still effective 7 days post dose and that the data across the 5 studies were homogenous (Fig. 1). This is a first step in validating the use of ketamine in Phase I development as a positive control for NMDA activity.

Figure 1: Forests plots from Lee et al. 2015 on Day 1 (left panel) and Day 7 (right panel). Numerous animal models have been used which showed the rapid antidepressant effect of ketamine10. Interestingly, the effect of ketamine was blocked by pre-treatment with rapamycine, thus confirming the involvement mTOR signaling in the antidepressant effect of ketamine.

2 Pain Applications

Apart from its use in psychiatry, ketamine as an NMDA antagonist prototype has been tested and used in several pain therapeutic protocols. It is used as an add-on to opioids or as a sparing agent in cancer pain or acute pain11,12. Ketamine also reduces morphine-induced hyperalgesia, through a mechanism mediated by the phosphorylation of the NMDA receptor linked to the activation of Protein Kinase C13,14. Many trials have also been conducted with or , however ketamine seems the most efficacious. NMDA antagonists also have the potential to provide release from neuropathic or orofacial pain, especially combined with opioids15,16 2003) even though clinical data is currently limited17.

Biomarkers and Surrogate Endpoints of NMDA Antagonist Activity in Non-Clinical and Early Clinical Development

By observing the effects of NMDA antagonist activity, as outlined above, it is possible to identify various biomarkers that could be of use, prior to planning a Phase IIb trial, in order to measure target engagement.

Methods exist for psychiatry and pain applications, several of which are translational (T):

• High amplitude spontaneous gamma oscillations and increased connectivity (T) • Mismatch Negativity (MMN) reduction (T), reduction of the P3a and P3b components in animals and of the P300 in humans (T) • Increase in neurotrophic factor BDNF plasma levels (T) • Clinical assessment of psychotomimetic effects (both negative and positive symptoms) and deficit in working memory • Shift of emotional processing with Facial Emotion Recognition Task (FERT) • Bold imaging of various regions in resting state • Wind-up, i.e. temporal summation of stimuli leading to pain in normal or sensitized skin (T) • Reversal of acutely induced remifentanil hyperalgesia (T) • Induced gamma produced either by auditory stimuli or visual stimuli. Both are up and running and have undergone a validation process inclusive of test-retest reliability (see Fig. 5) • Thermal grill illusion Biotrial has the ability to measure all of these biomarkers and is therefore perfectly positioned to assist clients with the early development of NMDA antagonist compounds. These models are up and running and some are compatible with the single and multiple dose study design combined with an add-on arm.

3 The Use of Ketamine as Positive Control in the Development of NMDA Antagonists

The reference drug ketamine has been extensively studied and a dosing regimen of a bolus + infusion was tested for its cognitive, psychometric and electrophysiological effects (Fig. 2).

Figure 2: Clinical data (Brief Psychiatric Rating Scale total and Scale for Assessment of Negative Symptoms) in 12 male subjects in a 4-way crossover study conducted at Forenap.

An application of this was the use of ketamine as a positive control in the development of lanicemine, as shown in Fig. 3, where the dissociative symptoms observed with ketamine were not observed with Lanicemine.

Figure 3: Effects of ketamine and lanicemine on Clinician Administered Dissociative State Scale (from Sanacora7 et al. 2015) conducted at Forenap.

Similarly, when qEEG was used in a study with lanicemine which also used ketamine as a positive control, the first step was to test the lack of nystagmus (involuntary eye movement) at the dose used, as nystagmus artificially creates gamma oscillations18. In this study19 there was no effect on optokinetic parameters and a dose-dependent increase in the gamma-1 and gamma-2 bands was observed. The magnitude of the ketamine effect can be achieved in the range between doses of 75 mg and 150 mg of lanicemine (Fig. 4). Induced gamma is also of relevance and a paradigm was set up and tested for its reproducibility (Fig. 5). In addition, Lanicemine was tested in MDD patients at a single dose of 100 mg, which induced a reduction in the blood oxygen level-dependent (BOLD)

4 signaling in the subgenual cingulate gyrus (NB subgenual cingulate and visual cortical responses to sad, but not happy, facial stimuli are correlated with changes in symptom score) and reduction of negatively valenced stimuli (angry face pictures) induced activation of the amygdala20 as an acute PD endpoint.

Figure 4: Effects in healthy subjects of ketamine and lanicemine19.

When tested in efficacy trials, dosing three times a week produced an effect7 while a single infusion of lanicemine failed to produce an antidepressant effect21. The sponsor made the strategic decision to halt CNS activities, which included halting the compound’s development, despite a promising profile.

Figure 5: Method validation for evoked gamma oscillation and resting gamma oscillation in healthy subjects.

5 In another study the effect of ketamine on mismatch negativity, a ketamine-sensitive biomarker, was in part reversed by 0.8 mg/kg, which binds at the co-agonist site19 (Fig. 6).

Figure 6: Partial reversal of the effects of ketamine on mismatch negativity by oral glycine in the healthy male subject.

Integrating Biomarkers and PD Models into Phase I Programs

In proposing a strategy for an early development program for an NMDA antagonist, we would recommend the use of adaptive CNS methodologies to investigate target engagement as well as pharmacological and clinical activity, in addition to standard safety, tolerability and pharmacokinetic assessments. When and where possible, these methods would be included in the Phase I Single (SAD) and Multiple (MAD) dose studies. However, for certain tests such as pain models or advanced CNS methods, SAD and MAD designs may not provide enough sensitivity or allow for comparison with ketamine. In such cases we would recommend the inclusion of an additional arm in an integrated protocol. A list of possibilities (non-exhaustive), per study type and indication, would be: First-in-Human Studies • Use of qEEG on a limited number of electrodes in the SAD study in an incomplete block design with rotating panels, all subjects receiving placebo once • Specific assessment of psychotomimetic effects balancing the small numbers by a high sensitivity • Self-administered rating scales

MAD Study • Use of qEEG on a limited number of electrodes in parallel group testing (only sensitive to high magnitude effects) • Assay of BDNF levels • Specific assessment of psychotomimetic effects balancing the small numbers by a high sensitivity • Self–assessments

Pharmacodynamic Study in a 4-Way Crossover • Two sub-MTD doses of the new compound compared, within subjects, to placebo and a ketamine infusion in 16 subjects. This single dose study could be run in parallel to the MAD. • qEEG repeated measurements with spectral analysis and connectivity analysis • Scales for psychotomimetic effects • General cognitive testing including working memory at peak

6 Pain Models

The pain models that can be used for the assessment of ketamine-like compounds are based on wind- up (“wind-up” is a frequency-dependent increase in the excitability of spinal cord neurons, evoked by electrical stimulation of afferent C-fibres), when the stimulus is repeated, which occurs after the glutamate release and the activation of the AMPA, leading to the activation of the NMDA receptor. Several methods are available for the measurement of the wind-up by mechanical repeated stimuli, using a Von Frey hair or with a temporal summation with electric discharges. Implementation of these tests in early development is among Biotrial’s areas of expertise. This includes a validated prototype for applying flare-inducing electrical stimulus in a needle-free manner22.

Figure 7: Flare and pain Visual Analogue Scale (VAS) during the application of the needle free transdermal stimulator. Maximum gradient if ion flux produces very small visible lesions that heal in 2-3 days.

Due to the stimulus specificity of response to ketamine, several tests would be used in a4-way crossover study with ketamine and a placebo control and two sub-MTD dose levels of the new compound in pain-phenotyped healthy male subjects: • Temporal summation of a Von Frey stimulation in primary and secondary hyperalgesia produced by a mild thermal burn23. Temporal summation from repeated TDES stimulation and secondary hyperalgesia and allodynia • Thermal grill illusion which has been shown to be sensitive to ketamine24 selectively on the difference between the two temperatures without modifying the perception or pain thresholds • Standard Quantitative Sensory Testing (brief testing) Modalities of testing and models are based on the animal models shown to be drug-sensitive (e.g. the UV erythema, capsaicin neuropathic pain models, etc.). Conclusion Tools exist to assist teams in their early development decision-making process, when looking to identify the most promising compounds in their portfolio. The addition of dedicated tests to Phase I programs is a cost-effective and time-saving way to gain precious knowledge on a compound. This can, in turn, be used in the go/no-go decision process as well as in planning the next steps of clinical development. Biotrial’s experience and expertise in designing and performing efficient programs for its clients is available to early clinical development teams looking to maximize their NMDA antagonist assets.

7 References

1) Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB Jr, Charney DS. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994 Mar;51(3):199-214. 2) Steriade M. Corticothalamic networks, oscillations, and plasticity. Adv Neurol. 1998;77:105-34. Review. 3) Destexhe A. Spike-and-wave oscillations based on the properties of GABAB receptors. J Neurosci. 1998 Nov 1;18(21):9099-111. 4) Pinault D. N-methyl d-aspartate receptor antagonists ketamine and MK-801 induce wake-related aberrant gamma oscillations in the rat neocortex. Biol Psychiatry. 2008 Apr 15;63(8):730-5. 5) Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, Krystal JH. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000 Feb 15;47(4):351-4. 6) Cook D, Brown D, Alexander R, March R, Morgan P, Satterthwaite G, Pangalos MN.Lessons learned from the fate of AstraZeneca’s drug pipeline: a five-dimensional framework. Nat Rev Drug Discov. 2014 Jun;13(6):419-31. 7) Sanacora G, Smith MA, Pathak S, Su HL, Boeijinga PH, McCarthy DJ, Quirk MC.Lanicemine: a low-trapping NMDA channel blocker produces sustained antidepressant efficacy with minimal psychotomimetic adverse effects. Mol Psychiatry. 2014 Sep;19(9):978-85. 8) Dale E, Bang-Andersen B, Sánchez C. Emerging mechanisms and treatments for depression beyond SSRIs and SNRIs. Biochem Pharmacol. 2015 Mar 24. pii: S0006-2952(15)00156-2. 9) Lee EE, Della Selva MP, Liu A, Himelhoch S. Ketamine as a novel treatment for major depressive disorder and bipolar depression: a systematic review and quantitative meta-analysis. Gen Hosp Psychiatry. 2015 Mar-Apr;37(2):178-84. 10) Browne CA, Lucki I. Antidepressant effects of ketamine: mechanisms underlying fast-acting novel . Front Pharmacol. 2013 Dec 27;4:161. 11) Bredlau AL, Thakur R, Korones DN, Dworkin RH. Ketamine for pain in adults and children with cancer: a systematic review and synthesis of the literature. PainMed. 2013 Oct;14(10):1505-17. 12) Bell RF, Eccleston C, Kalso EA. Ketamine as an adjuvant to opioids for cancer pain. Cochrane Database Syst Rev. 2012 Nov 14;11:CD003351. 13) Mao J, Price DD, Mayer DJ. Mechanisms of hyperalgesia and morphine tolerance: a current view of their possible interactions. Pain. 1995 Sep;62(3):259-74. 14) Zhao YL, Chen SR, Chen H, Pan HL. Chronic opioid potentiates presynaptic but impairs postsynaptic N-methyl-D- receptor activity in spinal cords: implications for opioid hyperalgesia and tolerance. J Biol Chem. 2012 Jul 20;287(30):25073-85. 15) Pelissier T, Laurido C, Kramer V, Hernández A, Paeile C. Antinociceptive interactions of ketamine with morphine or in mononeuropathic rats. Eur J Pharmacol. 2003 Sep 5;477(1):23-8. 16) Alvarez P, Saavedra G, Hernández A, Paeile C, Pelissier T. Synergistic antinociceptive effects of ketamine and morphine in the orofacial capsaicin test in the rat. Anesthesiology. 2003 Oct;99(4):969-75. 17) Marchetti F, Coutaux A, Bellanger A, Magneux C, Bourgeois P, Mion G. Efficacy and safety of oral ketamine for the relief of intractable chronic pain: Aretrospective 5-year study of 51 patients. Eur J Pain. 2014 Nov 10. doi:10.1002/ejp.624. [Epub ahead of print] PubMed PMID: 25381898. 18) Radant AD, Bowdle TA, Cowley DS, Kharasch ED, Roy-Byrne PP. Does ketamine-mediated N-methyl-D-aspartate receptor antagonism cause schizophrenia-like oculomotor abnormalities? Neuropsychopharmacology. 1998 Nov;19(5):434-44. 19) Boeijinga P, Danjou P., Patroneva A, Smith M & Quirk - Low-trapping NMDA channel blocker AZD6765 increases gamma-band EEG without dissociative side-effects: a comparison with ketamine in healthy volunteers ACNP Poster 2011.

8 20) Deakin JFW, Williams S,Downey D, McKie S, Goodwin G, Rylands A, Harmer C, Craig K & Dourish CT- PharmacoMRI and cognitive effects of the low-trapping NMDA channel blocker AZD6765 compared with ketamine in untreated major depressive disorder – CINP 2012 Poster P09-0.13. 21) Zarate CA Jr, Mathews D, Ibrahim L, Chaves JF, Marquardt C, Ukoh I, Jolkovsky L, Brutsche NE, Smith MA, Luckenbaugh DA. A randomized trial of a low-trapping nonselective N-methyl-D-aspartate channel blocker in major depression. Biol Psychiatry. 2013 Aug 15;74(4):257-64. 22) Lecybyl R, Acosta J, Ghoshdastidar J, Stringfellow K, Hanna M. Validation, reproducibility and safety of trans dermal electrical stimulation in chronic pain patients and healthy volunteers. BMC Neurol. 2010 Jan 13;10:5. 23) Schulte H, Sollevi A, Segerdahl M. The synergistic effect of combined treatment with systemic ketamine and morphine on experimentally induced windup-like pain in humans. Anesth Analg. 2004 Jun;98(6):1574- 80. 24) Kern D, Pelle-Lancien E, Luce V, Bouhassira D. Pharmacological dissection of the paradoxical pain induced by a thermal grill. Pain. 2008 Apr;135(3):291-9.

9