QurAlis Announces Topline Data From its Phase 1 Clinical Trial of QRL-101 Evaluating Biomarkers of ALS and Epilepsy in Healthy Volunteers
Results demonstrated statistically significant effects on biomarkers that predict ALS disease progression and severity
Evidence of brain penetration, target engagement, and potential anti-seizure effects demonstrated through biomarkers for TMS-EMG and EEG
Safety and tolerability profile consistent with previously reported results for QRL-101
QurAlis intends to advance QRL-101 into proof-of-concept studies as a potentially best-in-class treatment for both ALS and epilepsy
CAMBRIDGE, Mass., March 12, 2025 /PRNewswire/ -- QurAlis Corporation ('QurAlis'), a clinical-stage biotechnology company driving scientific breakthroughs into powerful precision medicines that have the potential to alter the trajectory of neurodegenerative and neurological diseases, today announced positive topline data from QurAlis' Phase 1 proof-of-mechanism (PoM) clinical trial of QRL-101 in healthy volunteers evaluating biomarkers related to amyotrophic lateral sclerosis (ALS) and epilepsy.
Topline results demonstrated a dose-dependent, statistically significant decrease in motor nerve excitability threshold tracking (mNETT) strength-duration time constant (SDTC), the co-primary endpoint of the trial related to ALS. SDTC is a well-established electrophysiological biomarker of peripheral motor excitability related to ALS, where elevation of SDTC has been clinically shown to predict faster disease progression and mortality. The decrease in SDTC observed with QRL-101 in this study was approximately 50% greater than previously reported for the Kv7 potassium channel opener ezogabine in a single-dose study in people with ALS. Additional observations for multiple secondary and exploratory endpoints related to motor nerve excitability were also significantly impacted.
The clinical trial demonstrated a statistically significant impact on multiple secondary and exploratory endpoints related to epilepsy. These included transcranial magnetic stimulation electromyography (TMS-EMG) intracortical facilitation which indicates effective inhibition of cortical excitability, as well as significant increases in electroencephalography (EEG) spectral power in beta and gamma spectral bands further demonstrating cortical target engagement and brain penetration. There was minimal to no impact on the slower delta and theta EEG bands, which suggests a low potential for GABA-A receptor activation and sedation. The study did not reach statistical significance on the co-primary endpoint of TMS-EMG motor evoked potential (MEP) amplitude, another measure of corticospinal excitability.
Results also demonstrated that the safety and tolerability profile of QRL-101 was consistent with previously reported study results evaluating QRL-101 to date. There were no serious adverse events or discontinuations due to adverse events reported observed in the study.
'We are excited by these topline data from our biomarker study in healthy participants which suggest that QRL-101 has the potential to provide a therapeutic effect for both ALS and epilepsy,' said Kasper Roet, Ph.D., CEO and co-founder of QurAlis. 'Loss of Kv7.2/7.3 function from the mis-splicing of the KCNQ2 gene leading to hyperexcitability-induced neurodegeneration was one of the first genetic breakthroughs from human ALS motor neuron stem cell models made by our founders at Harvard. QurAlis was started to develop a best-in-class Kv7 ion channel opener for the treatment of hyperexcitability-induced disease progression in ALS, which occurs in about half of all people living with ALS. Elevated SDTC has been clinically linked to faster disease progression and mortality, underscoring the need for an effective treatment for this population.'
Dr. Roet continued, 'The decrease in SDTC observed with QRL-101 in this study was approximately 50% greater than previously reported for the Kv7 opener ezogabine in a single-dose study in people with ALS. QRL-101 has the potential to be a promising therapeutic, especially considering the encouraging results from a previous study with ezogabine in ALS patients showing positive effects on both SDTC and the disease progression biomarker compound muscle action potential (CMAP). Further, based on the strong clinical validation of Kv7.2/7.3 ion channel openers in seizure reduction, the TMS-EMG intracortical facilitation together with the EEG data from this study suggest QRL-101 may have the potential to reduce both frequency and severity of seizures in people living with epilepsy without causing sedation. We are encouraged by these results and look forward to advancing the clinical program for QRL-101 in both ALS and epilepsy so that we can fulfill our mission of bringing much-needed precision medicine options to patients.'
Kv7.2/7.3 is a voltage-gated potassium channel whose role is crucial for the regulation of both neuronal excitability and membrane potential. QRL-101 is a potentially best-in-class selective Kv7.2/7.3 ion channel opener for the treatment of hyperexcitability-induced disease progression in ALS, which occurs in both sporadic and genetic forms of ALS, with the majority caused by the mis-splicing of the KCNQ2 gene in the pre-mRNA. Kv7 is also a clinically validated target to regulate the hyperexcitable state in epilepsy. In vivo and in vitro studies have demonstrated that QRL-101 is more potent in threshold track studies and exhibits the potential for fewer clinical adverse events that ezogabine, a less selective, first-generation, Kv7.2/7.3 channel opener.
The Phase 1 PoM study (QRL-101-05, NCT06681441) was a randomized, double-blind, placebo-controlled, three-way crossover trial. The study was conducted at the Centre for Human Drug Research, an early-phase Contract Research Organization based in Leiden, The Netherlands. The study evaluated two dose levels of QRL-101 versus placebo in healthy volunteers, assessing:
Additional QRL-101 clinical trials include:
QRL-101-01 (NCT05667779) was a first-in-human, single-ascending dose clinical trial in 88 healthy participants. In this clinical trial, QRL-101 was shown to be well tolerated. The study completed in late 2023 and results from QRL-101-01 supported a tolerable dose range for subsequent studies.
QRL-101-03 (NCT06532396) is a randomized, double-blind, placebo-controlled, multiple-ascending dose clinical trial evaluating the safety, tolerability, and PK of QRL-101 in up to 60 healthy participants.
QRL-101-04 (NCT06714396) is a Phase 1 PoM single-dose, placebo-controlled clinical trial designed to evaluate the safety and tolerability of QRL-101 in people living with ALS. QRL-101-04 is expected to enroll approximately 12 participants with ALS and will evaluate the impact of QRL-101 on mNETT.
QRL-101-06 is a Phase 1 randomized, open-label, single dose, cross-over study to evaluate the PK of three formulations of QRL-101 in a fasted condition or in the presence of a high fat meal in healthy participants.
More information about the QRL-101 clinical trials can be found at www.clinicaltrials.gov.
About QurAlis Corporation
At QurAlis, we are neuro pioneers on a quest to cure, boldly seeking to translate scientific breakthroughs into powerful precision medicines. We work collaboratively with a relentless pursuit of knowledge, precise attention to craft, and compassion to discover and develop medicines that have the potential to transform the lives of people living with neurodegenerative and neurological diseases. QurAlis is the leader in development of precision therapies for amyotrophic lateral sclerosis (ALS). In addition to ALS, QurAlis is advancing a robust precision medicine pipeline to bring effective disease-modifying therapeutics to patients suffering from severe diseases defined by genetics and clinical biomarkers. For more information, please visit www.quralis.com or follow us on X @QurAlisCo or LinkedIn.

Try Our AI Features
Explore what Daily8 AI can do for you:
Comments
No comments yet...
Related Articles
Yahoo
30 minutes ago
- Yahoo
MMA INNOVATE VIETNAM 2025: Important Marketing Event for Vietnam Martech Ecosystem
HO CHI MINH CITY, Vietnam, June 11, 2025 /PRNewswire/ -- MMA INNOVATE VIETNAM, the country's pioneering platform shaping the future of marketing, is back on 21 August 2025 — bigger, bolder, and brimming with opportunities to connect, collaborate, and co-create the next wave of digital transformation. The Vietnam MarTech Report 2024 shows just how quickly the space is evolving: the number of MarTech companies has skyrocketed from 15–20 players in 2023 to nearly 100 in 2024. Furthermore, the KPMG 2024 Customer Experience Excellence Report which highlights how Vietnamese businesses are already leveraging AI to elevate trust, security, and emotional connection — and you've got the perfect storm for innovation. Thus, INNOVATE 2025's theme, "Shaping Growth Through Marketing Insight, Impact, and Innovation," is more than a slogan. It's a call to action for top brands, ambitious SMEs, startups, and marketers to come together and thrive in the age of AI, MarTech, and experience-driven landscape of AI, MarTech, and customer experience. Impressive Numbers from MMA INNOVATE VIETNAM 2024 2,500+ delegates from all corners of the marketing industry Over 30 exhibition booths featuring major names like Samsung, Heineken, Coca-Cola, Nestlé, Unilever, Suntory PepsiCo, AIA, AdFlex, Ecomobi, Haravan, AnyMind, MGID, VCCorp, VietGuys, Chicilon Media, Novaon, Ureka Media, Appier, and many more. 62 speakers from multi-nation corps delivering global insights 25,000+ live views via MMA's social channels and media partners 20 million+ total reach across online and offline platforms Over 300 B2B connections made directly at the event. "MMA INNOVATE VIETNAM 2025 is not just an event – it's a launchpad for businesses to thrive. We believe AI and MarTech are the keys to creating meaningful differentiation for sustainable growth. MMA Global Vietnam is committed to supporting the business community, helping them seize opportunities, overcome challenges, and accelerate in the digital era." – Ms. Phan Bich Tam, Country Director, MMA Global Vietnam. Who Should Exhibit? Businesses and startups in marketing, MarTech, advertising, media, data, AI, and e-commerce. Providers of digital transformation solutions, technology platforms, and creative services. Brands looking to launch new products/services and seeking strategic partners. Book Your Booth Don't miss the chance to connect and grow your business at MMA INNOVATE VIETNAM 2025! Contact the organizing team via email: 0937 811 381Limited booths available – Book early to secure the best spots! PR Newswire is the official Media Partner of the MMA INNOVATE VIETNAM 2025. View original content to download multimedia: SOURCE MMA Vietnam Sign in to access your portfolio
Yahoo
30 minutes ago
- Yahoo
Kanazawa University research: High-speed AFM and 3D modelling help towards understanding the dynamics of a protein implicated in several cancers
KANAZAWA, Japan, June 11, 2025 /PRNewswire/ -- Researchers at the Nano Life Science Institute (WPI-NanoLSI), Kanazawa University observe and model how the enzyme ADAR1 interacts with double-stranded RNA, which may be useful for future cancer treatment strategies. An enzyme type noted in several cancers is the family of adenosine deaminases acting on RNA (ADARs). These enzymes convert adenosines in double-stranded RNA (dsRNA) into inosines, which cells read as guanosines. As such, ADARs can contribute to changes in protein-coding sequences and diminish the robustness of various RNA processes. Studies have shown that silencing one type of ADAR – ADAR1 – can prevent cancer proliferation and sensitize cancers to immunotherapy, suggesting that they could be a promising target for cancer treatments. However, so far, it has been difficult to pin down information on the structural dynamics of ADAR1 due to its size and complexity. Now, researchers led by Madhu Biyani at Kanazawa University, WPI-NanoLSI, Yasuhiro Isogai at Toyama Prefectural University, and Manish Biyani at Ishikawa Create Labo and Kwansei Gakuin University have combined high-speed atomic force microscopy (HS-AFM) and 3D modelling to shed light on the enzyme's conformations and interactions with dsRNA. Like many proteins, ADAR1 functions through changes in its conformation. However, most experimental techniques for determining protein structure, as well as 3D modelling algorithms, give static or average conformations that obscure the structural dynamics so important to the protein's function. Combining 3D modelling with HS-AFM proved helpful in shedding light on these dynamic aspects of ADAR1. The researchers first used 3D modelling based on the machine learning algorithm AlphaFold2 to predict the conformations of the enzyme and noted that it could take the form of monomers, dimers, trimers, and tetramers. HS AFM observations, as well as theoretically simulated HS AFM, supported these initial conclusions regarding the possible oligomer formations. The researchers then looked at the conformations the enzyme formed in the presence of double-stranded RNA (dsRNA). In particular, the researchers focused on a certain aryl hydrocarbon receptor 3'UTR mRNA as the target for ADAR1, since this receptor is known to be involved in the metabolism of substances alien to the body at those points. Observations of the dsRNA with HS-AFM not only agreed with previous structural studies but were able to provide insights into the structure of the target region in particular. Thanks to the speed and resolution of the HS-AFM image capture, the researchers were able to identify different conformations in the proteins that seemed to relate to distinct phases of the deaminizing process. In their report of the work, the researchers explain how ADAR1 first "searches" for the dsRNA and on "recognizing" it, adopts a flexible conformation as it approaches. The enzyme then engages in what the researchers describe as "capture" of the dsRNA backbone, for which the conformation transitions to something more stable and "anchor-like". The researchers highlight the role of dsRNA binding domains (dsRBDs) to stabilize the interaction with the dsRNA at this stage. They also note "a visibly large interfacial interaction between the deaminase domains, forming a dimer" as the enzyme dimer loops out on the dsRNA. The enzyme subsequently scans the RNA and dissociates to search for adenosine sites to convert. "These observations suggest that the dsRBDs are critical for initiating interactions between the deaminase domains, thereby promoting the formation of a stable, functional dimeric complex capable of efficiently binding and catalyzing the editing of dsRNA substrates," the researchers conclude in their report, thereby flagging the insights this study offers for further work towards possible cancer therapeutics. The researchers further propose future studies to compare ADAR1 and ADAR2, and to perform mutation analyses to clarify how ADAR1 dimerization influences A-to-I RNA editing, ultimately aiming to develop effective ADAR1 inhibitors. Figure 1: Three-dimensional modeling analysis of ADAR1 2025 Biyani, et al., Nature Communications Figure 2: Simulated AFM images of ADAR1 2025 Biyani, et al., Nature Communications Glossary Atomic force microscopy This imaging technique uses a nanosized tip at the end of a cantilever that is scanned over a sample. It can be used to determine the topography of a sample surface from the change in the strength of forces between the tip and the sample with distance, and the resulting deflection of the cantilever. It was first developed in the 1980s, but a number of modifications have augmented the functionality of the technique since. It is better suited to imaging biological samples than the scanning tunnelling microscope that had been previously developed because it does not require a conducting sample. In the 2000s, Toshio Ando at Kanazawa University was able to improve the scanning speed to such an extent that moving images could be captured. This allowed people to use the technique to visualize dynamic molecular processes for the first time. ADARs Adenosine deaminases acting on RNA (ADARs) convert adenosine to inosine, which is interpreted by cells as guanosine. This editing can influence alternative splicing, miRNA processing, double-stranded RNA stability, and protein-coding sequences. In mammals, there are two known ADARs responsible for adenosine to inosine RNA editing – ADAR1 and ADAR2. Notably, overexpression and increased activity of ADAR1 have been observed in cancers of the liver, breast, esophagus, prostate, and bone marrow. 3D modelling Several techniques are now available for protein structure prediction. In this study, the researchers used AlphaFold2 to model the structure of human ADAR1. Due to disorder regions in the N-terminal 822 residues, the final model focused on residues 823-1226, which encompass the deaminase domain. This monomer model served as the basis for building higher-order structures—dimer, trimer, tetramer, and polymer by superimposing it onto the ADAR2 dimer structure (PDB ID:1ZY7) through sequence alignment. Since determining ADAR1's full structure is challenging due to its size and complexity, the better-characterized ADAR2 provided a template to generate ADAR1 multimer models. Reference Madhu Biyani, Yasuhiro Isogai, Kirti Sharma, Shoei Maeda, Hinako Akashi, Yui Sugai, Masataka Nakano, Noriyuki Kodera, Manish Biyani, Miki Nakajima, High-speed atomic force microscopy and 3D modeling reveal the structural dynamics of ADAR1 complexes, Nature Communications 16, 4757 (2025). DOI:10.1038/s41467-025-59987-6 URL: Funding and AcknowledgementsFinancial support from the Grants-in-Aid for Scientific Research (C), KAKENHI, Japan Society for the Promotion of Science (JSPS) (23K06067 to MadhuB), and the World Premier International Research Center Initiative (WPI), MEXT, Japan, are gratefully acknowledged. The authors thank Prof. Toshio Ando, Dr. Kenichi Umeda, Ms. Wei Weilin, Ms. Aimi Makino, and Ms. Kayo Nagatani for their technical support of HS-AFM experiments. Contact Kimie Nishimura (Ms)Project Planning and Outreach, NanoLSI Administration OfficeNano Life Science Institute, Kanazawa UniversityEmail: nanolsi-office@ Kanazawa 920-1192, Japan About Nano Life Science Institute (WPI-NanoLSI), Kanazawa UniversityUnderstanding nanoscale mechanisms of life phenomena by exploring "uncharted nano-realms". Cells are the basic units of almost all life forms. We are developing nanoprobe technologies that allow direct imaging, analysis, and manipulation of the behavior and dynamics of important macromolecules in living organisms, such as proteins and nucleic acids, at the surface and interior of cells. We aim at acquiring a fundamental understanding of the various life phenomena at the About the World Premier International Research Center Initiative (WPI)The WPI program was launched in 2007 by Japan's Ministry of Education, Culture, Sports, Science and Technology (MEXT) to foster globally visible research centers boasting the highest standards and outstanding research environments. Numbering more than a dozen and operating at institutions throughout the country, these centers are given a high degree of autonomy, allowing them to engage in innovative modes of management and research. The program is administered by the Japan Society for the Promotion of Science (JSPS). See the latest research news from the centers at the WPI News Portal: WPI program site: About Kanazawa UniversityAs the leading comprehensive university on the Sea of Japan coast, Kanazawa University has contributed greatly to higher education and academic research in Japan since it was founded in 1949. The University has three colleges and 17 schools offering courses in subjects that include medicine, computer engineering, and humanities. The University is located on the coast of the Sea of Japan in Kanazawa, a city rich in history and culture. The city of Kanazawa has a highly respected intellectual profile since the time of the fiefdom (1598-1867). Kanazawa University is divided into two main campuses: Kakuma and Takaramachi for its approximately 10,200 students, including 600 from View original content: SOURCE Kanazawa University
Yahoo
an hour ago
- Yahoo
Energenesis Biomedical to Unveil Promising Parkinson's Disease Therapy, ENERGI-F705PD, at BIO International 2025
TAIPEI, June 11, 2025 /PRNewswire/ -- Energenesis Biomedical Co., Ltd. (TWSE: 6657), a clinical-stage biotechnology company pioneering cellular energy restoration therapeutics, today announced its participation in the BIO International Convention 2025, taking place June 16–19 in Boston, MA. The company will be located at Booth #1645 within the Taiwan Pavilion, to showcase its innovative ENERGI Platform and provide a key update on its investigational Parkinson's disease (PD) therapy, ENERGI-F705PD, a first-in-class treatment with disease-modifying potential. Key Highlights: ENERGI-F705PD is a first-in-class, oral sustained-release therapy in development for Parkinson's disease, designed to address the underlying pathogenesis. Topline data from the Phase I trial in healthy subjects are anticipated in September 2025, following successful completion of recruitment. ENERGI-F705PD aims to combat the root causes of Parkinson's by reducing alpha-synuclein aggregation, enhancing antioxidant capacity, and boosting dopamine synthesis. "We are eager to present the exciting progress of our ENERGI-F705PD program at BIO International 2025," said Dr. Han-Min Chen, CEO of Energenesis Biomedical. "Parkinson's disease remains a challenging condition with significant unmet needs. Our approach focuses on restoring cellular energy and combating critical pathogenesis of PD, offering a potential disease-modifying solution for patients. We look forward to sharing our latest data and exploring potential partnerships that can accelerate the development of this important therapy." About ENERGI-F705PD ENERGI-F705PD is a novel small-molecule drug delivered in an oral sustained-release formulation. It is designed to enhance cellular ATP and antioxidant production by engaging the purine salvage, glycolysis, and pentose phosphate pathways. This multi-pronged mechanism of action directly addresses key pathological hallmarks of Parkinson's disease: Reduce and Prevent α-Synuclein Aggregation: Elevated ATP acts as a hydrotrope, helping prevent the intracellular aggregation of misfolded α-synuclein, a central pathogenic hallmark of PD. Increase Reducing Power (antioxidant): ENERGI-F705PD upregulates the pentose phosphate pathway (PPP), leading to increased NADPH production and enhanced cellular antioxidant defense. Increase Tyrosine Hydroxylase Expression/activity: By optimizing cellular energy metabolism and engaging the purine salvage pathway, ENERGI-F705PD also enhances the expression and activity of tyrosine hydroxylase (TH), the key enzyme in dopamine synthesis, thereby helping restore neurotransmitter levels in the brains of PD patients. About Energenesis Biomedical Energenesis Biomedical Co., Ltd. (TWSE: 6657) is a clinical-stage biopharmaceutical company dedicated to developing innovative therapeutics that restore cellular energy and activate intrinsic repair mechanisms. By combining proprietary small-molecule innovation with AI-driven drug repurposing, Energenesis Biomedical aims to accelerate the development of treatments for unmet medical needs across various therapeutic areas, including neurodegenerative diseases, chronic wounds, and rare disorders. The company's robust pipeline includes: ENERGI-F705PD: Oral tablet for Parkinson's disease (Phase I) ENERGI-F703DFU: Gel for Diabetic foot ulcers (Phase III) ENERGI-F701: Tonic for Alopecia (Phase II completed) ENERGI-F703EB: Cream for Epidermolysis bullosa (EB), granted FDA Orphan Drug and Rare Pediatric Disease designation, and EMA orphan designation(Preparing for Phase II) For more information, please visit Contact: Business Development Team Email: partnership@ View original content: SOURCE ENERGENESIS BIOMEDICAL CO., LTD. Error in retrieving data Sign in to access your portfolio Error in retrieving data Error in retrieving data Error in retrieving data Error in retrieving data