8-K
false000175523700017552372022-06-102022-06-10

 

UNITED STATES

SECURITIES AND EXCHANGE COMMISSION

Washington, D.C. 20549

 

 

 

 

 

 

FORM 8-K

 

 

 

 

 

 

CURRENT REPORT

Pursuant to Section 13 or 15(d)

of the Securities Exchange Act of 1934

 

Date of Report (Date of earliest event reported): June 10, 2022

 

 

 

 

 

 

CYCLERION THERAPEUTICS, INC.

(Exact name of registrant as specified in its charter)

 

 

 

 

 

 

Massachusetts

 

001-38787

 

83-1895370

(State or other jurisdiction

of incorporation)

 

(Commission

File Number)

 

(IRS Employer

Identification Number)

 

245 First Street, 18th Floor

Cambridge, Massachusetts 02142

(Address of principal executive offices, including Zip Code)

 

Registrant’s telephone number, including area code: (857) 327-8778

 

Check the appropriate box below if the Form 8-K filing is intended to simultaneously satisfy the filing obligation of the registrant under any of the following provisions:

 

Written communications pursuant to Rule 425 under the Securities Act (17 CFR 230.425)

 

Soliciting material pursuant to Rule 14a-12 under the Exchange Act (17 CFR 240.14a-12)

 

Pre-commencement communications pursuant to Rule 14d-2(b) under the Exchange Act (17 CFR 240.14d-2(b))

 

Pre-commencement communications pursuant to Rule 13e-4(c) under the Exchange Act (17 CFR 240.13e-4(c))

 

Securities registered pursuant to Section 12(b) of the Act:

 

Title of each class

 

Trading Symbol(s)

 

Name of each exchange on which registered

Common Stock, no par value

 

CYCN

 

The Nasdaq Stock Market LLC

(Nasdaq Global Select Market)

 

Indicate by check mark whether the registrant is an emerging growth company as defined in Rule 405 of the Securities Act of 1933 (§230.405 of this chapter) or Rule 12b-2 of the Securities Exchange Act of 1934 (§240.12b-2 of this chapter). Emerging growth company

 

If an emerging growth company, indicate by check mark if the registrant has elected not to use the extended transition period for complying with any new or revised financial accounting standards provided pursuant to Section 13(a) of the Exchange Act.

 


 

Item 7.01 Regulation FD Disclosure.

On June 10, 2022, Cyclerion Therapeutics, Inc. (the "Company") announced topline data from its CY6463 Mitochondrial Encephalomyopathy Lactic Acidosis and Stroke-like episodes (”MELAS”) study. Copies of the press release and corporate presentation are being furnished as Exhibit 99.1 and Exhibit 99.2, respectively to this Current Report on Form 8-K.

The information contained in Item 7.01 of this Current Report on Form 8-K, Exhibit 99.1 and Exhibit 99.2 attached hereto is intended to be furnished and shall not be deemed “filed” for purposes of Section 18 of the Securities Exchange Act of 1934 (the “Exchange Act”) or otherwise subject to the liabilities of that section, nor shall it be deemed incorporated by reference in any filing under the Securities Act of 1933, as amended, or the Exchange Act, except as expressly set forth by specific reference in such filing.

 

Item 9.01 Financial Statements and Exhibits.

 

(d)

 

Exhibit No.

 

Description

 

 

 

99.1

 

Press Release of Cyclerion Therapeutics, Inc. dated June 10, 2022

99.2

 

Corporate presentation of Cyclerion Therapeutics, Inc., dated June 10, 2022

104

 

Cover Page Interactive Data File

 

2


 

SIGNATURES

 

Pursuant to the requirements of the Securities Exchange Act of 1934, the registrant has duly caused this report to be signed on its behalf by the undersigned hereunto duly authorized.

 

 

Cyclerion Therapeutics, Inc.

 

 

 

 

 

 

Dated: June 10, 2022

By:

/s/ Anjeza Gjino

 

 

Name:

Anjeza Gjino

 

 

Title:

Chief Financial Officer

 

3


EX-99.1

Exhibit 99.1

https://cdn.kscope.io/47e91ea65e306f678d40a04b66202ed0-img121346916_0.jpg 

 

 

Cyclerion Therapeutics Announces Positive Topline Clinical Data for CY6463 in MELAS Patients at UMDF Mitochondrial Medicine 2022 Symposium

 

Data from an eight-patient, open-label study demonstrate improvements across multiple biomarkers of mitochondrial function, inflammation, cerebral blood flow, and functional connectivity

 

CY6463 was well tolerated, with no reports of serious adverse events (SAEs) or treatment discontinuation due to adverse events (AEs); oral, once-daily administration provided expected CNS exposure

 

Data support further development of CY6463 in CNS diseases with mitochondrial dysfunction

 

Cambridge, Mass., June 10, 2022 – Cyclerion Therapeutics, Inc. (Nasdaq: CYCN), a clinical-stage biopharmaceutical company on a mission to develop treatments that restore cognitive function, today announced positive topline data in its signal-seeking clinical study of CY6463, for the potential treatment of Mitochondrial Encephalomyopathy, Lactic Acidosis and Stroke-like episodes (MELAS). Chad Glasser, Pharm.D., Director of Clinical Research at Cyclerion Therapeutics, will present results from this clinical study today during the Clinical Trial Updates Panel at the United Mitochondrial Disease Foundation (UMDF) Mitochondrial Medicine 2022 Symposium, taking place June 8-11, 2022, in Phoenix, Arizona.

 

CY6463 is a positive allosteric modulator of soluble guanylate cyclase (sGC), which amplifies endogenous NO signaling, a pathway that has been linked to mitochondrial biogenesis and function. In this open-label, single-arm study of the oral, once-daily sGC stimulator in eight MELAS patients, improvements were seen across a range of biomarkers, including mitochondrial disease-associated biomarkers such as lactate and GDF-15, a broad panel of inflammatory biomarkers, cerebral blood flow, and functional connectivity between neural networks. These positive effects after 29 days of dosing were supported by correlations across several endpoints and were more pronounced in patients with greater baseline disease burden. A return toward baseline levels after discontinuation of CY6463 dosing across several biomarkers was also observed.

 


CY6463 was well tolerated with no adverse events leading to treatment discontinuation, and pharmacokinetics (PK) were consistent with the Phase 1 study in healthy volunteers. The positive data from this study further support the potential of CY6463, the first and only CNS-penetrant sGC stimulator in clinical development, to provide therapeutic benefit to people living with MELAS.

 

“MELAS patients currently have no approved treatment options for a devastating orphan disease that affects multiple organs, including the CNS, skeletal muscle, and eyes,” said Peter Hecht, Ph.D., Chief Executive Officer of Cyclerion. “We are excited by the strength of these data and consistency across disease domains, which support the further advancement of CY6463 as a potential treatment option.”

 

Study Highlights:

The single-arm, open-label study enrolled eight participants who spanned a range of disease burden; 6 of the 8 (75%) were also taking a daily regimen of oral arginine or citrulline, precursors to nitric oxide that are current standard of care for MELAS patients.
CY6463 was well tolerated; there were no reports of serious adverse events (SAEs) or treatment discontinuation due to adverse events (AEs).
The PK profile and concentrations in the cerebrospinal fluid (CSF) and plasma were consistent with exposures observed in Phase 1 healthy volunteer studies.
Effects were observed across multiple domains of disease activity:
o
Improvements in biomarkers associated with mitochondrial function including lactate and GDF-15. These changes correlated with each other and with CY6463 plasma concentrations
o
Improvements across a broad panel of inflammatory biomarkers
o
Increases in cerebral blood flow across all brain regions. These changes correlated with clinical improvement as assessed by the patient global impression of change (PGIC) scale
o
Increases in functional connectivity between brain regions and activation of occipital brain regions in response to the visual stimulus as measured by fMRI BOLD

“In this study we saw positive impacts on important biomarkers associated with MELAS and other mitochondrial disease following 29 days of once-daily dosing with CY6463,” said Andreas Busch, Ph.D., Chief Scientific Officer at Cyclerion Therapeutics. “These findings are exciting as we think


about the potential of our mechanism in mitochondrial disease and more broadly about the effects of CY6463 on mitochondrial function, which is relevant to numerous CNS diseases, including schizophrenia and Alzheimer’s Disease.”

 

A video presentation of the topline data is available on the Investor page of the Cyclerion website. Additional data from the MELAS clinical study will be shared in the coming weeks.

 

About CY6463

CY6463 is the first CNS-penetrant sGC stimulator to be developed as a symptomatic and potentially disease-modifying therapy for serious CNS diseases. The nitric oxide (NO)-soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP) signaling pathway is a fundamental mechanism that precisely controls key aspects of physiology throughout the body. In the CNS, the NO-sGC-cGMP pathway regulates diverse and critical biological functions including neuronal function, neuroinflammation, cellular bioenergetics, and vascular dynamics. Although it has been successfully targeted with several drugs in the periphery, this mechanism has yet to be fully leveraged therapeutically in the CNS, where impaired NO-sGC-cGMP signaling is believed to play an important role in the pathogenesis of many neurodegenerative and neuropsychiatric diseases and other disorders associated with cognitive impairment. As an sGC stimulator, CY6463 acts as a positive allosteric modulator to sensitize the sGC enzyme to NO, increase the production of cGMP, and thereby amplify endogenous NO signaling. By compensating for deficient NO-sGC-cGMP signaling, CY6463 and other sGC stimulators may have broad therapeutic potential as a treatment to improve cognition and function in people with serious CNS diseases.

 

About the Study

The Phase 2a study was an open-label, single-arm study of oral, once-daily CY6463 in eight adults aged 18 or older with MELAS. The primary objective of the study was to assess the safety and tolerability of a 15 milligram, once-daily, oral dose of CY6463 over 29 days. The secondary objectives included pharmacokinetics, and exploratory pharmacodynamic effects, with the goal of identifying which biomarkers to carry forward into additional studies. The study was not powered for hypothesis testing.

 

About MELAS

Mitochondrial Encephalomyopathy, Lactic Acidosis, and Stroke-like episodes (MELAS) is a devastating orphan disease affecting multiple organ systems, including the CNS, with no approved


therapies. It is the most common form of primary mitochondrial diseases (PMD). MELAS is phenotypically and genetically defined by a mutation in mitochondrial tRNA. It is estimated that about 1 in 4,300 individuals has a mitochondrial disease, and ~80% of individuals with mitochondrial disease have CNS symptoms. The unmet need in MELAS is immense, symptoms include, chronic fatigue, muscle weakness, and pain in addition to neurological manifestations. Life expectancy is estimated at ~17 years from onset of CNS symptoms. The disease impedes the individual’s ability to live independently, leads to social isolation, and overall reduced quality of life.

 

About Cyclerion Therapeutics
Cyclerion Therapeutics is a clinical-stage biopharmaceutical company on a mission to develop treatments that restore cognitive function. Cyclerion is advancing novel, first-in-class, CNS-penetrant, sGC stimulators that modulate a key node in a fundamental CNS signaling pathway. The multidimensional pharmacology elicited by the stimulation of sGC has the potential to impact a broad range of CNS diseases. The most advanced compound, CY6463, has shown rapid improvement in biomarkers associated with cognitive function and is currently in clinical development for Mitochondrial Encephalomyopathy, Lactic Acidosis and Stroke-like episodes (MELAS), Cognitive Impairment Associated with Schizophrenia (CIAS) and Alzheimer's Disease with Vascular pathology (ADv). Cyclerion is also advancing CY3018, a next-generation sGC stimulator.

 

Forward Looking Statement

Certain matters discussed in this press release are “forward-looking statements”. We may, in some cases, use terms such as “predicts,” “believes,” “potential,” “continue,” “estimates,” “anticipates,” “expects,” “plans,” “intends,” “may,” “could,” “might,” “will,” “should”, “positive” or other words that convey uncertainty of future events or outcomes to identify these forward-looking statements. In particular, the Company’s statements regarding trends and potential future results are examples of such forward-looking statements. The forward-looking statements include risks and uncertainties, including, but not limited to, the success, timing and cost of our ongoing or future clinical trials and anticipated clinical trials for our current product candidates, including statements regarding the timing of initiation and completion of the trials, futility analyses and receipt of interim results, which are not necessarily indicative of or supported by the final results of our ongoing or subsequent clinical trials; any results of clinical studies, including in particular single-arm open-label studies involving a number of patients that is not statistically significant such as described in this release, not necessarily being indicative of or supported by the final results of our ongoing or subsequent clinical trials; our ability to fund additional clinical trials to continue the advancement of our product candidates; the timing of and our ability to obtain and maintain U.S. Food and Drug Administration (“FDA”) or other regulatory


authority approval of, or other action with respect to, our product candidates; the potential for the CY6463 clinical trial to provide a basis for approval for treatment of MELAS; the Company’s ability to successfully defend its intellectual property or obtain necessary licenses at a cost acceptable to the Company, if at all; the successful implementation of the Company’s research and development programs and collaborations; the success of the Company’s license agreements; the acceptance by the market of the Company’s product candidates, if approved; and other factors, including general economic conditions and regulatory developments, not within the Company’s control. The factors discussed herein could cause actual results and developments to be materially different from those expressed in or implied by such statements. The forward-looking statements are made only as of the date of this press release and the Company undertakes no obligation to publicly update such forward-looking statements to reflect subsequent events or circumstance.

 

For more information about Cyclerion, please visit cyclerion.com and follow us on Twitter and LinkedIn.

 

Investors

Carlo Tanzi, Ph.D.

Kendall Investor Relations

ctanzi@kendallir.com

 

Media

Amanda Sellers

Verge Scientific Communications

asellers@vergescientific.com

 


Slide 1

Thinking differently about cognition CORPORATE Presentation June 2022 Exhibit 99.2


Slide 2

Safe harbor statement This presentation is for informational purposes only and is not an offer to sell nor a solicitation of an offer to buy any securities of Cyclerion Therapeutics, Inc. (the “Company”). This presentation includes or may include certain information obtained from trade and statistical services or sources, third party publications and other sources. The Company has not independently verified such information and there can be no assurance as to its accuracy. Certain matters discussed in this presentation are “forward-looking statements”. We may, in some cases, use terms such as “predicts,” “believes,” “potential,” “continue,” “estimates,” “anticipates,” “expects,” “plans,” “intends,” “may,” “could,” “might,” “will,” “positive,” “should” or other words that convey uncertainty of future events or outcomes to identify these forward-looking statements. In particular, the Company’s statements regarding trends and potential future results are examples of such forward-looking statements. The forward-looking statements include risks and uncertainties, including, but not limited to, the success, timing and cost of our ongoing or future clinical trials and anticipated clinical trials for our current product candidates, including statements regarding the timing of initiation and completion of the trials, futility analyses and receipt of interim results, which are not necessarily indicative of or supported by the final results of our ongoing or subsequent clinical trials; any results of clinical studies, including in particular single-arm open-label studies involving a number of patients that is not statistically significant such as described in this presentation, not necessarily being indicative of or supported by the final results of our ongoing or subsequent clinical trials; our ability to fund additional clinical trials to continue the advancement of our product candidates; the timing of and our ability to obtain and maintain U.S. Food and Drug Administration (“FDA”) or other regulatory authority approval of, or other action with respect to, our product candidates; the potential for the CY6463 clinical trial to provide a basis for approval for treatment of MELAS; the Company’s ability to successfully defend its intellectual property or obtain necessary licenses at a cost acceptable to the Company, if at all; the successful implementation of the Company’s research and development programs and collaborations; the success of the Company’s license agreements; the acceptance by the market of the Company’s product candidates, if approved; and other factors, including general economic conditions and regulatory developments, not within the Company’s control. The factors discussed herein could cause actual results and developments to be materially different from those expressed in or implied by such statements. The forward-looking statements are made only as of the date of this presentation and the Company undertakes no obligation to publicly update such forward-looking statements to reflect subsequent events or circumstance.   Other important factors that could cause actual results to differ from those reflected in any forward-looking statements herein are described in the Company’s most recent Form 10-K as well as the Company’s subsequent filings with the Securities and Exchange Commission (the “SEC”). All of the Company’s development plans may be subject to adjustment depending on funding, recruitment rate, regulatory review, preclinical and clinical results, and other factors any of which could result in changes to the Company’s development plans and programs or delay the initiation, enrollment, completion, or reporting of clinical trials.   In addition to the risks described above and in the Company’s filings with the SEC, other unknown or unpredictable factors could affect the Company’s results. No froward-looking statements can be guaranteed, and actual results may materially differ from such statements. The information in this presentation is provided only as of June 10, 2022, and the Company undertakes no obligation to update any forward-looking statements contained in this presentation on account of new information, future events, or otherwise, except as required by law.


Slide 3

Developing therapies to restore cognitive function Experienced team of scientific leaders, drug hunters, company builders, and CNS experts Developed and launched first-in-class therapies Raised billions in capital Built leading strategic partnerships Neuroinnovation engine enables faster, more precise drug development Advancing CY6463, potential breakthrough, first-in-class CNS therapy  Improvement trends in MELAS patients across biomarkers of mitochondrial function, inflammation, cerebral blood flow and functional connectivity CIAS study results expected in Q3’22; ADv study actively enrolling Evaluating collaborative development opportunities Developing disease and mechanism specific translational biomarkers Continuously refining signal-to-noise Identifying most promising patient populations early


Slide 4

Neuroinnovation Engine


Slide 5

Efficiently developing drugs that matter Identify most promising patient populations early, increase signal-to-noise ratio Leverage deep understanding of brain biology, pathobiology of cognitive dysfunction and small molecule mechanisms Partner with academic and industry leaders to access leading-edge data and technologies Apply advanced analytics to robust multimodal nonclinical and clinical data sets to extract actionable insights


Slide 6

MOA & biomarker-driven non-clinical validation Identify mechanism- and disease-relevant biomarkers Proof of principle; first-in-human biomarker translation Validate homologous physiological response to drive clinical development Proof of mechanism; biomarker-guided, signal-seeking studies in patients Refine indication and patient selection aided by advanced analytics Proof of concept; Accelerate and de-risk Inform larger, longer studies Late-stage development and approval Select the right patient, right dose, right endpoint Scientific insights & unmet need Cyclerion neuroinnovation engine de-risks development at every stage Insights & Analytics


Slide 7

Deploying our NeuroInnovation ENGINE


Slide 8

DISCOVERY IND-ENABLING PHASE 1* PHASE 1b/2a PHASE 2 CY6463 MELAS CIAS ADv Multiple under assessment CY3018 Multiple under assessment Advancing parallel clinical studies in priority populations *Two phase 1 studies were completed in healthy young and old (>65 years of age) volunteers confirming targeted CNS exposure and activity


Slide 9

CY6463 amplifies the fundamental NO-sGC-cGMP signaling pathway CY6463 First-in-class BBB-permeable, positive allosteric modulator of sGC Amplifies endogenous NO-sGC-cGMP signaling to address central aspects of disease pathophysiology sGC stimulators clinically validated in several non-CNS indications Preclinical data and extensive academic work validate the crucial role of the NO-sGC-cGMP pathway in brain physiology Important role in learning and memory Scientific insights & unmet need MOA & biomarker-driven non-clinical validation First-in-human biomarker translation Biomarker-guided, signal-seeking studies in patients Mitochondrial Function Inflammatory Processes Cerebral Blood Flow Neuronal Function PKG, PDE, ion channels Downstream targets (eg, CREB, BDNF)


Slide 10

CY6463 improves processes relevant to cognition in preclinical models Mitochondrial Function Cerebral Blood Flow Inflammatory Processes Neuronal/Cognitive Function Reticular activating system Thalamus Cortical transition areas Ventral hippocampus Young rat/Vehicle Aged rat/Vehicle Aged rat/CY6463 Scientific insights & unmet need MOA & biomarker-driven non-clinical validation First-in-human biomarker translation Biomarker-guided, signal-seeking studies in patients


Slide 11

CY6463 showed rapid improvement in biomarkers associated with cognitive function Increased alpha and gamma power Improved N200 latency Alpha power: CY6463 vs. placebo Day 15 Improvement Age (years) Untreated CY6463 treated Time (ms) Reduced neuroinflammatory biomarkers In a 15-day study in 24 healthy elderly subjects, CY6463 demonstrated: Scientific insights & unmet need MOA & biomarker-driven non-clinical validation First-in-human biomarker translation Biomarker-guided, signal-seeking studies in patients


Slide 12

Biomarker-guided strategy to refine target populations with cognitive impairment ADv | Alzheimer’s Disease with vascular pathology (ADv)  CIAS | Cognitive Impairment Associated with Schizophrenia MELAS | Mitochondrial Encephalomyopathy, Lactic Acidosis, and Stroke-like episodes ADv MELAS Mitochondrial Disease Parallel studies in distinct populations Efficient, signal-seeking studies inform larger and longer studies Disease-relevant biomarkers accelerate and guide development Improving Cognition Neuropsychiatric CIAS Neurodegenerative Significant additional opportunities Translation of insights across programs increases odds of success Scientific insights & unmet need MOA & biomarker-driven non-clinical validation First-in-human biomarker translation Biomarker-guided, signal-seeking studies in patients


Slide 13

MELAS Study Results


Slide 14

MELAS clinical data demonstrate that CY6463 has potential as breakthrough CNS therapeutic MELAS Devastating genetically and phenotypically defined mitochondrial disease (MD) with no approved therapies CY6463 Positive allosteric modulator of sGC which amplifies endogenous NO signaling Study design Open label, 29-day study of once-daily, oral, CY6463 (n=8) Improvement observed across important biomarkers associated with MELAS after CY6463 treatment Well tolerated with no serious adverse events or treatment discontinuation Oral once-daily administration provided expected CNS exposure  Improvements observed across multiple domains of disease activity: Biomarkers associated with mitochondrial function, including lactate and GDF-15 Broad panel of inflammatory biomarkers with the potential to translate to CNS diseases with mitochondrial dysfunction Cerebral blood flow (CBF) across all brain regions Functional connectivity between brain regions and activation of occipital brain regions in response to the visual stimulus as measured by fMRI BOLD Supported by correlations across several endpoints and more pronounced in patients with heavier baseline disease burden Return toward baseline levels observed across several biomarkers after dosing discontinuation


Slide 15

MELAS*: devastating orphan disease affecting multiple organ systems, no approved therapies  Multisystem involvement Skeletal muscle Muscle weakness Myopathy Exercise intolerance Eyes Vision abnormalities Orphan disease MELAS is most common form of primary mitochondrial disease (PMD) Phenotypically and genetically defined (mutation in mitochondrial tRNA) ~10-20k MELAS patients (US) ~65k PMD patients (US) Tremendous unmet need Life expectancy ~17 years from onset of CNS symptoms Chronic fatigue, muscle weakness, and pain in addition to neurological manifestations Impedes ability to live independently Social isolation, and reduced quality of life >80% of patients have CNS symptoms Nervous system Stroke-like episodes Cognitive impairment Seizures Headaches Ataxia Hearing loss Altered consciousness Peripheral neuropathy *MELAS: Mitochondrial Encephalomyopathy, Lactic Acidosis, & Stroke-like Episodes


Slide 16

Strong therapeutic rationale for stimulating NO-sGC-cGMP pathway to treat mitochondrial disease CY6463 is a positive allosteric modulator of sGC and amplifies endogenous NO signaling Literature links NO-sGC-cGMP pathway to mitochondrial biogenesis and function NO deficiency in mitochondrial disease has been linked to impaired blood flow, inflammation, angiopathy, and endothelial dysfunction Use of NO precursors recommended by Mitochondrial Medicine Society CYCN preclinical data demonstrate CY6463 affects  multiple aspects of mitochondrial disease pathophysiology


Slide 17

Open-label, 29-day study of CY6463 in MELAS patients to assess safety, PK, PD and impact on important domains of mitochondrial disease CNS/PD Safety Safety and tolerability profile with 15-mg QD dosing Safety on top of NO precursors and other stable medications  PK Study population (n=8) Genetically confirmed with history of CNS symptoms such as stroke, seizure, headache Stable medications including NO precursors (e.g., arginine and citrulline) permitted (6 of 8) Plasma and, when available, cerebrospinal fluid (CSF) concentrations of CY6463 Measures of key domains of MELAS Mitochondrial function Inflammatory processes Cerebral blood flow Neuronal/cognitive function Patient-reported outcomes (PROs)


Slide 18

Strong safety/tolerability and once-daily profile extended to participants with MELAS CY6463 well tolerated with and without NO precursors (L-arginine and L-citrulline) Mostly mild adverse events (AEs), no severe AEs No SAEs, no discontinuations due to AEs Most common AE was headache, all but 1 mild No signals on clinical labs, vital signs, ECGs, or suicidal rating scale Once-daily dosing with consistent pharmacokinetics Pharmacokinetics (AUCtau, Cmax, and Ctrough) in MELAS participants consistent with PK studies in healthy volunteers Confirmed CNS exposure with CSF:plasma ratio consistent with that observed in healthy volunteers


Slide 19

Lactate (mmol/L)  (effect size = -0.5) GDF-15 (ng/mL)  (effect size = -0.5) FGF-21 (ng/mL) (effect size = -0.2) Improvement in biomarkers of mitochondrial function that are affected in MELAS disease Mitochondrial function effects Blood biomarkers linked to mitochondrial function were elevated at baseline across participants (mean) Improvement after 29-day dosing supported by correlations between blood biomarkers and CY6463 plasma concentrations Mean Change from Baseline and 90% CI improvement GDF-15: Growth/Differentiation Factor-15; FGF-21: Fibroblast Growth Factor-21


Slide 20

B2M (μg/mL) (effect size = -1.0) SAP (μg/mL) (effect size = -0.9) Broad improvement of inflammatory biomarkers  Anti-inflammatory effects ~65% of 40 inflammatory biomarkers with effect sizes |value|≥0.3 Central and peripheral inflammation is upregulated in patients with mitochondrial dysfunction TNFR2 (ng/mL) (effect size = -0.7) Mean Change from Baseline and 90% CI Biomarkers with effect size |value|≥0.7 improvement B2M: Beta-2-Microglobulin; SAP: Serum Amyloid P-Component; TNFR2: Tumor Necrosis Factor Receptor 2


Slide 21

Increased cerebral blood flow across all regions analyzed Blood flow effects Neuronal and/or glial injury due to mitochondrial failure, nitric oxide deficiency and cerebrovascular angiopathy reduce cerebral blood flow Dysregulated cerebral blood flow is linked to stroke-like episodes and CNS symptoms (mL/100g/min) Whole Brain Bilateral (effect size = 0.3) Temporal Lobe Bilateral (effect size = 0.4) Parietal Lobe Bilateral (effect size = 0.2) Occipital Lobe Bilateral (effect size = 0.3) Frontal Lobe Bilateral (effect size = 0.4) Corpus Callosum Bilateral (effect size = 0.4) Cingulate Lobe Bilateral (effect size = 0.4) Cerebral White Matter Bilateral (effect size = 0.3) Cerebellar White Matter Bilateral (effect size = 0.4) Mean change from baseline and 90% CI improvement


Slide 22

CY6463 enhanced functional connectivity and visual activation in CNS, which is impaired in MELAS Task-free fMRI (resting state) shows enhanced functional connectivity: Increased signals across several resting state networks including those involved in:   executive function sensorimotor processing Task-based fMRI (visual activation) shows occipital region activation by CY6463 fMRI BOLD response to visual stimulus is markedly reduced in symptomatic MELAS compared to controls (Rodan et al 2020) CY6463 increased activation of occipital brain regions in response to the visual stimulus, with greater activation at Day 29 compared to screening and Day 1 Additional analyses of imaging data ongoing n=6 (fMRI data collected at one site were not analyzable) Whole-brain voxelwise statistical parametric maps (SPM) of task-fMRI visual activation at day 1 (green) and day 29 (red) visits compared to screening. Maps thresholded a t = 2.0 for exploratory visualization. Day 1 (green) and Day 29 (red) visits compared to screening Task-based fMRI visual activation


Slide 23

Improvements after 29-day dosing supported by correlation across endpoints Correlations (r) between CBF and clinical improvement as assessed by the patient global impression of change (PGIC) Correlations (r) between changes in blood biomarkers and plasma concentrations Biomarker parameters Fibroblast growth factor 21 Growth differentiation factor 15 Lactate Trough plasma concentration Fibroblast growth factor 21 1.00 Growth differentiation factor 15 0.86 1.00 Lactate 0.74 0.87 1.00 Trough plasma concentration -0.75 -0.68 -0.41 1.00 Darker greens are correlations ≥0.8 (very strong) Lighter greens are correlations ≥0.6 but <0.8 (strong)


Slide 24

Ongoing clinical trials


Slide 25

CIAS study ongoing; data expected Q3 2022 Objectives Exploratory, signal-seeking study to evaluate safety, tolerability, and pharmacodynamic effects (qEEG, ERP, digital cognitive performance battery) Study design In-clinic, randomized, placebo-controlled, double-blind, multiple-ascending-dose design 14-day treatment with Once-daily CY6463 or placebo 48 participants across 4 sequential cohorts Patient targeting Psychiatrically stable adults with schizophrenia, no more than moderate symptoms On stable, single antipsychotic regimen Collaborations Study conducted at experienced, partner sites: Hassman Research Institute and Collaborative Neuroscience Exploratory, AI-driven, integrated analysis of data with Ariana Pharma Scientific insights & unmet need MOA & biomarker-driven non-clinical validation First-in-human biomarker translation Biomarker-guided, signal-seeking studies in patients


Slide 26

ADv study ongoing Exploratory, signal-seeking study to evaluate safety, tolerability, and pharmacodynamic effects (EEG, MRI, neuroinflammatory biomarkers, cognition) Once-daily CY6463 vs. placebo 12 weeks 30 participants Confirmed AD pathology (PET, CSF) 2+ cardiovascular risk factors Mild-moderate subcortical small-vessel disease on MRI Mini mental state exam score (20-26) Objectives Study design Patient targeting Partially funded by the Alzheimer's Association’s Part the Cloud-Gates Partnership Collaborating with Dr. Andrew Budson at Boston University on a study to examine the relationship between ERP/EEG and cognitive measures in dementias  Collaborations Scientific insights & unmet need MOA & biomarker-driven non-clinical validation First-in-human biomarker translation Biomarker-guided, signal-seeking studies in patients


Slide 27

Next-generation sGC stimulator program


Slide 28

Applying the neuroinnovation engine to CY3018 – a differentiated sGC stimulator In mice, rats, and non-human primates, CY3018 has greater partitioning into the brain than CY6463 CY3018 elicited unique patterns of activation and deactivation in rodent imaging studies demonstrating differentiation from other sGC stimulators IND-enabling activities are on track for end-of-year completion Greater relative CNS exposure Greater relative CNS pharmacology Scientific insights & unmet need MOA & biomarker-driven non-clinical validation Demonstrated a positive effect on cognition in primates  Currently exploring the impact of CY3018 in preclinical models Expected to be a once-daily oral therapy


Slide 29

Track record of success


Slide 30

Leadership Team with Track Record of Success Peter Hecht, PhD Chief Executive Officer Andy Busch, PhD Chief Scientific Officer  Chris Winrow, PhD  Vice President, Translational Medicine & Development Program Lead Jennifer Chickering, PhD Vice President, Clinical Strategy Anjeza Gjino, MBA Chief Financial Officer Cheryl Gault Chief Operating Officer Todd Milne, PhD  Sr. Vice President, Corporate Development Bill Kissel, PhD Vice President, Pharmaceutical Development Bruce Kinon, MD Vice President, Clinical Development Kevin Durfee Vice President, Information Technology & Facilities


Slide 31

George Conrades Errol De Souza, PhD Marsha Fanucci, Chair Peter Hecht, PhD, CEO Ole Isacson, MD, PhD Stephanie Lovell Terrance McGuire Michael Mendelsohn, MD Experienced Board of Directors and Scientific Advisors Board of Directors Scientific Advisors Claudio Babiloni, PhD University of Rome  Andrew E. Budson, MD VA Boston Healthcare System Boston University Harvard Medical School Mark Currie, PhD Ironwood Pharmaceuticals Science Exchange Marni J. Falk, MD The Children’s Hospital of Philadelphia University of Pennsylvania Michael Heneka, MD, PhD Luxembourg Centre for Systems Biology  University of Massachusetts Robert C. Malenka, MD, PhD Nancy Pritzker Laboratory Stanford University David H. Salat, PhD MGH / Harvard Medical School VA Boston Healthcare System Daniela Salvemini, PhD St. Louis University Harald H.H.W. Schmidt, MD, PhD, PharmD Maastricht University Eric Smith, MD University of Calgary M Brandon Westover, MD, PhD MGH / Harvard Medical School Beacon Biosignals Chris Wright, MD, PhD AavantiBio


Slide 32

Developing therapies to restore cognitive function Experienced team of scientific leaders, drug hunters, company builders, and CNS experts Developed and launched first-in-class therapies Raised billions in capital Built leading strategic partnerships Neuroinnovation engine enables faster, more precise drug development Advancing CY6463, potential breakthrough, first-in-class CNS therapy  Improvement trends in MELAS patients across biomarkers of mitochondrial function, inflammation, cerebral blood flow and functional connectivity CIAS study results expected in Q3’22; ADv study actively enrolling Evaluating collaborative development opportunities Developing disease and mechanism specific translational biomarkers Continuously refining signal-to-noise Identifying most promising patient populations early


Slide 33

Appendices Preclinical, Phase 1 and translational pharmacology studies, references


Slide 34

Preclinical Data


Slide 35

CY6463 demonstrated beneficial effects in preclinical studies across multiple domains associated with cognitive disease Cerebral Blood Flow Increased blood flow in areas associated with memory and arousal by fMRI BOLD imaging IMPROVED Cellular Bioenergetics Increased ATP and restored gene expression in cells from patients with mitochondrial diseases ENHANCED Neuronal Function Enhanced memory & spine density in aged animals; increased LTP in neurodegenerative models; affected qEEG spectra IMPROVED Neuro- inflammation Decreased markers of LPS-induced neuroinflammation (ICAM1, VCAM1, IL6) in vitro REDUCED


Slide 36

CY6463 improved neuronal function Restored hippocampal long-term potentiation to wild-type levels in a mouse neurodegenerative model Wild type Disease Disease + CY6463 (46 nM) * Normalized fEPSP Amplitude Time (min) 10 30 50 70 1.0 5 2.0 2.5 Improve Neuronal Function Reduce Neuroinflammation Enhance Cellular Bioenergetics Improve Cerebral Blood Flow Hippocampal slices from symptomatic Huntington’s Disease (R6/2) mice incubated with CY6463 for 25-30 minutes before LTP induction Extracellular field potentials recordings performed using Multi-Electrode Array; **p<0.01 vs. Disease By acting directly on the neurons, CY6463 could restore impaired neurotransmission


Slide 37

CY6463 increased qEEG gamma power No effect seen with PDE9 inhibitor Healthy awake rats were treated with clinically relevant doses of CY6463 (3 mg/kg) or PDE9 inhibitor (10 mg/kg) Graph displays 1-2h post-dose, mean ± SEM Improve Neuronal Function Reduce Neuroinflammation Enhance Cellular Bioenergetics Improve Cerebral Blood Flow CY6463 is differentiated from PDE9 inhibitor, which showed no effect on gamma power Gamma Power Vehicle PDE9i CY6463 CY6463 + PDE9i


Slide 38

CY6463 and donepezil act independently to enhance qEEG signal Combination elicited additive increase in gamma band power in healthy rats Improve Neuronal Function Reduce Neuroinflammation Enhance Cellular Bioenergetics Improve Cerebral Blood Flow *p<0.05 vs Veh # p<0.05 CY6463 vs CY6463 +Donepezil Healthy rats orally administered CY6463 (10mg/kg), Donepezil (1mg/kg), or a combination. Graph displays 1-2h post-dose, mean ± SEM CY6463 may offer opportunity to enhance attention and cognitive performance alone and on top of standard of care Gamma Power Vehicle Donepezil CY6463 CY6463 + Donepezil


Slide 39

CY6463 improved learning and memory in aged rats Increased rate of learning in aged rats treated with CY6463 in Morris Water Maze Healthy aged male rats were administered CY6463 (10 mg/kg, p.o.) daily during Morris Water Maze training *p<0.05 vs. Aged vehicle-treated Improve Neuronal Function Reduce Neuroinflammation Enhance Cellular Bioenergetics Improve Cerebral Blood Flow


Slide 40

CY6463 improved cognitive function in pharmacologically impaired rats Improve Neuronal Function Reduce Neuroinflammation Enhance Cellular Bioenergetics Improve Cerebral Blood Flow Rat Novel Object Recognition *p<0.05 vs. VEH + MK-801 rats CY6463 acts downstream of NMDA receptor to reverse deficit induced by NMDA antagonist (MK-801)


Slide 41

Mushroom spine density Restoration of spine density has potential to provide neuroprotective effects and improve synaptic function in neurodegenerative diseases CY6463 improved neuronal function Enhanced hippocampal spine density in aged animals treated with CY6463 Young Control Aged Control Aged CY6463 Control Young CY6463 Control Aged * * Improve Neuronal Function Reduce Neuroinflammation Enhance Cellular Bioenergetics Improve Cerebral Blood Flow *p<0.05 vs. Aged 3-month old (young) or 16-month old (aged) healthy mice at study initiation Aged mice treated for 4 months with 1 mg/kg CY6463


Slide 42

CY6463 reduced neuroinflammation Inhibited in vitro LPS-induction of biomarkers of neuroinflammation *p<0.05 vs. control LPS-treated wells CY6463 (10 µM) and DETA (30 µM) were incubated with SIM-A9 cells or rat brain 3D microtissues for 30 minutes before LPS (100 ng/ml) incubation and further incubated for 18-20h at 37oC before IL-6 quantification in the media Neuroinflammation in mouse microglial cells Neuroinflammation in rat brain 3D microtissues * Improve Neuronal Function Reduce Neuroinflammation Enhance Cellular Bioenergetics Improve Cerebral Blood Flow CY6463 Control Control CY6463 Control Control * *


Slide 43

CY6463 enhanced cellular bioenergetics Increased ATP and restored decreased gene expression in cells from patients with mitochondrial diseases *p<0.05 vs. vehicle-treated wells GM13740 Leigh Syndrome patient cells obtained from the Coriell Institute were treated for 24h before ATP quantification TFAM: mitochondrial transcriptional factor A, a key activator of mitochondrial transcription as well as a participant in mitochondrial genome replication. Improve Neuronal Function Reduce Neuroinflammation Enhance Cellular Bioenergetics Improve Cerebral Blood Flow TFAM Mitochondrial disease patient cells * * * *


Slide 44

CY6463 improved cerebral blood flow Increased blood flow in areas associated with memory and arousal by fMRI BOLD imaging Peripherally restricted sGC stimulator CNS-penetrant sGC stimulator CY6463 Improve Neuronal Function Reduce Neuroinflammation Enhance Cellular Bioenergetics Improve Cerebral Blood Flow Healthy awake male rats treated with 0.3 mg/kg iv; image quantification 20-30 minutes post-dose


Slide 45

Phase 1 study results


Slide 46

Results CY6463 phase 1 showed CNS exposure, target engagement, PK, and safety PHASE 1 (completed Jan. 2020) 110 healthy young Age range 18-63 Standard safety Identified safe and well-tolerated dose levels with steady-state CNS exposure in therapeutic target range* Linear, predictable PK, consistent with QD dosing CNS exposure confirmed Evidence of target engagement (blood pressure) Goals Achieved Study design Three stages: SAD MAD Food Interaction PK (blood & CSF) Wide dose range tested Well tolerated at all dose levels, no safety signals May be taken with or without food *Based on positive CNS pharmacology in multiple preclinical models


Slide 47

Translational Pharmacology Study Results


Slide 48

CY6463 showed rapid and persistent improvements in multiple independent biomarkers associated with cognitive impairment In a 15-day study in 24 healthy elderly subjects CY6463 demonstrated: increased alpha and gamma power improved N200 latency faster saccadic eye movement (SEM) reaction time reduction in neuroinflammatory biomarkers Rapid onset (<15 days) Effect increased with age Biomarkers linked to AD and aging


Slide 49

Biomarker overview: qEEG frequency bands and their clinical implications qEEG is quantitative electroencephalography, an objective method that measures electrical activity and brain wave patterns Resting-state qEEG: subjects sit facing a featureless wall without moving recorded with eyes open and closed for 5 minutes each Band Frequency Hz associated with Delta 0-4 Deep sleep Theta 4-8 Waking/falling asleep, some with cognition Alpha 8-14 Passive wakefulness Attention and cognitive processing Beta 14-30 Alert, concentration Gamma 30-80 Higher cognitive function Associated with: Cognitive decline in aging and AD Genetic risk factors for AD (ApoE4) AD pathological protein levels (Aβ, tau) Improvement with approved AD treatments


Slide 50

CY6463 improved qEEG measures: significant increase in alpha power qEEG is quantitative electroencephalography, an objective method that measures electrical activity and brain wave patterns CY6463 vs. baseline Significant increase in EEG alpha power No effect of placebo Placebo vs. baseline CY6463 vs. placebo change (%) in alpha power on day 15


Slide 51

CY6463’s consistent alpha power effects across repeat sessions indicate stable and robust signal Footer DAY 1 baseline DAY 15 change from baseline CY6463 relative to placebo CY6463 Placebo Pre- dose 1 Pre- dose 2 Pre-dose last dose 2 hr post-dose 3 hr post-dose 6 hr post-dose Magnitude of improvement equivalent to decline seen after 2 years of aging


Slide 52

CY6463 increased alpha power with high responder rate (>70%) 1. Includes all subjects. For CY6463 and pbo each: n=12 for period 1, n=6 for period 2 Increase in alpha power Day 15 change from baseline in mean closed-eye alpha (8-12 Hz) Power Consistent individual treatment responses Posterior Closed-Eye Alpha (8-12 Hz) Power Placebo CY6463 Placebo CY6463 Posterior p = 0.0197 % 17% treatment effect over placebo Similar increase in anterior alpha power observed (p=0.0752) 13/18 participants increase with CY6463, vs 5/18 with placebo1 Overall effect not driven by outliers Placebo CY6463 BL BL D15 D15


Slide 53

CY6463 treatment associated with trend improvement in gamma power Change in Closed-Eye Gamma (25-45 Hz) Power Placebo CY6463 Placebo CY6463 200 150 100 50 0 -50 -100 Anterior (p = 0.081) Posterior (p = 0.1163) Change from baseline (%)


Slide 54

Biomarker overview: event-related potential (ERP) Trial: 500 tones 80% standard, 20% deviant Deviant Standard ERP oddball paradigm Subjects wear EEG cap and headphones, hear tones with instruction to press a button upon deviant tones P300 N200 N200 Stable component of ERP waveform Stimulus identification and distinction Affected in aging, neurodegenerative and neuropsychiatric diseases with cognitive impairment, and other CNS diseases Parameters Latency: time after the stimulus to peak signal Amplitude: size of peak signal


Slide 55

CY6463 improved N200 latency and effect increased with age Day 15 Improvement Age (years) Untreated CY6463 treated Time (ms) Effect of age on N200 latency Overall decrease in N200 latency for CY6463 treated vs untreated on day 15 (p<0.02) Effect more pronounced in older subjects


Slide 56

65 to 69y n=10 ≥70 y n=14 p=0.016 CY6463 improved N200 latency, driven by response in older subjects Greater decrease in ≥70y vs <70y Day 15 change from baseline Latency response was greater in subjects ≥70y vs 65-69y (p=0.016) Narrowing of variance in ≥ 70y supports a drug effect In ≥ 70y, magnitude of improvement after 2 weeks of treatment with CY6463 represents ~10y age-related change in N200 latency Time (ms) 3.0 1.5 0.0 1.5


Slide 57

Biomarker overview: saccadic eye movement as an objective measure of attention and cognition Short, fast, simultaneous tracking of both eyes in the same direction Brain areas involved include the frontal cortex, superior colliculus, substantia nigra, and amygdala Considered to be reflective of attention / arousal and influenced by motivation, time on task, and task difficulty Sensitive to sedation, fatigue, and CNS depressants and cognitive enhancers, and is affected by aging Peak Velocity Amplitude Latency Duration T=0ms Eye Position Eye Velocity https://www.liverpool.ac.uk/~pcknox/teaching/Eymovs/params.htm


Slide 58

CY6463 improved saccadic eye movement, an objective functional measure Decrease in saccadic reaction time Increase in saccadic peak velocity Shorter saccadic reaction times and faster saccadic velocities indicate that CY6463 is improving CNS functional performance – motor output – in addition to CNS neurophysiology Cognitive enhancers (e.g., modafinil) also positively impact saccadic eye movements Time (sec) p=0.0216 Placebo CY6463 Placebo CY6463 p=0.0216 p=0.07 Improvement Velocity (deg/sec) Improvement Mean change from baseline on day 15 post-dose 0.02 0.00 -0.02 -0.04 150 100 50 0


Slide 59

CY6463 improved neuroinflammatory biomarkers A2M and C3 are associated with pathological aging and Alzheimer’s Disease Alpha-2-macroglobulin (A2M) levels predict cognitive decline and development of AD; may lead to tau hyperphosphorylation Complement C3 (C3) colocalizes with Aβ plaques and tau tangles; involved in synaptic remodeling and degeneration LS % Mean Difference from placebo at Day 15 (95% CI)


Slide 60

Relevant reference publications


Slide 61

Relevant reference publications (1 of 2) NO-sGC-cGMP signaling in the CNS Garthwaite, John. “Nitric oxide as a multimodal brain transmitter.” Brain and neuroscience advances vol. 2 2398212818810683. 4 Dec. 2018  Kleppisch T, Feil R. cGMP signaling in the mammalian brain: role in synaptic plasticity and behaviour. Handb Exp Pharmacol. 2009;(191):549-79 Ben Aissa M, Lee SH, Bennett BM, Thatcher GR. Targeting NO/cGMP Signaling in the CNS for Neurodegeneration and Alzheimer's Disease. Curr Med Chem. 2016;23(24):2770-2788 Hollas MA, Ben Aissa M, Lee SH, Gordon-Blake JM, Thatcher GRJ. Pharmacological manipulation of cGMP and NO/cGMP in CNS drug discovery. Nitric Oxide. 2019 Jan 1;82:59-74 qEEG spectral frequency analysis Ishii et al. Healthy and Pathological Brain Aging: From the Perspective of Oscillations, Functional Connectivity, and Signal Complexity. Neuropsychobiology, 2018 Babiloni, et al. Resting-state posterior alpha rhythms are abnormal in subjective memory complaint seniors with preclinical Alzheimer's neuropathology and high education level: the INSIGHT-preAD study. Neurobiol Aging. 2020;90:43-59


Slide 62

Relevant reference publications (2 of 2) Event-related potential (ERP): MMN, N200 and P300 Bennys K, Portet F, Touchon J. Diagnostic value of event-related evoked potentials N200 and P300 subcomponents in early diagnosis of Alzheimer’s disease and mild cognitive impairment. J Clin Neurophysiol 2007;24:405–12 Fruehwirt et al. Associations of event-related brain potentials and Alzheimer’s disease severity: A longitudinal study. Progress in Neuropsychopharmacology and Biological Psychiatry 92 (2019) 31-38 Saccadic eye movement (SEM) Wilcockson et al. Abnormalities of saccadic eye movements in dementia due to Alzheimer’s disease and mild cognitive impairment. Aging 2019, Vol.11, No.15 James A. Sharpe & David H. Zackon (1987) Senescent Saccades: Effects of Aging on Their Accuracy, Latency and Velocity, Acta Oto-Laryngologica, 104:5-6, 422-428 ADv Cortes-Canteli M, Iadecola C. Alzheimer’s Disease and Vascular Aging: JACC Focus Seminar. J Am Coll Cardiol. 2020;75(8):942-951 MELAS El-Hattab AW, Adesina AM, Jones J, Scaglia F. MELAS syndrome: Clinical manifestations, pathogenesis, and treatment options. Mol Genet Metab. 2015;116(1-2):4-12 CIAS Keefe RS, Harvey PD. Cognitive impairment in schizophrenia. Handb Exp Pharmacol. 2012;(213):11-37


Slide 63