Author: Rachael Hansford

Ipsen at the World Congress for Neurorehabilitation

Ipsen had a robust presence at the 12th World Congress for Neurorehabilitation (WCNR) in Vienna, Austria and virtually, from 14-17 December 2022, further demonstrating the company’s long-standing partnership with the World Federation for Neurorehabilitation (WFNR) and ongoing commitment to neuroscience.

In addition to showcasing the company’s latest neuroscience clinical data, Ipsen hosted a satellite symposium entitled “About time: Optimising Long-Term Outcomes for Patients Treated with Botulinum Toxin”. Facilitated by global neuroscience experts Dr. Jorge Jacinto, Dr. Klemens Fheodoroff and Dr. Alberto Esquenazi, the session centred on new clinical data and around Ipsen’s patient-centric approach to educational training and the importance of focusing on long-term outcomes for patients treated with a botulinum toxin.

Data presented at WCNR 2022 included:

  1. A Phase IV, Prospective, Observational, Multicentre Study Evaluating the Effectiveness and Safety of abobotulinumtoxinA in Pediatric Lower Limb Spasticity (PLLS).
  2. A study exploring abobotulinumtoxinA efficacy and safety in children with upper limb spasticity previously treated with botulinum toxin.
  3. A study examining the change in Leg Activity Measure (LegA) sensitivity following treatment of lower limb spasticity with a single cycle of abobotulinumtoxinA in an ambulant population.
  4. Single centre, prospective, observational study methodology to assess the use of GripAble, a rehabilitation device, on patients with upper limb spasticity (ULS) receiving botulinum toxin A (BoNT-A).
  5. Baseline data from the AboLiSh study, exploring the most frequently injected muscles in patients undergoing treatment with abobotulinumtoxinA for lower limb spasticity in routine practice.
  6. The detrimental impact of the COVID-19 pandemic on the management of post-stroke spasticity in England.

Continuing to advance neuroscience and neurotoxin research

With over 25 years of experience in neurotoxins, neuroscience innovation remains central to the company’s future. Key data from Ipsen’s research and development programme reinforcing the company’s commitment to improving health outcomes for patients living with debilitating neurological disorders were at the WCNR. Clinical updates include a focus on the need to ensure treatment for spasticity is individualised, assessment of the impact of treatment with Dysport® (abobotulinumtoxinA) in pediatric and adult spasticity and a review of the impact of COVID-19 on stroke admissions and interventions in the UK.

“The breadth of data presented at the congress demonstrates our commitment to continued scientific innovation in neuroscience as well our focus on looking at patient care holistically to add value beyond our treatments. This aligns to our broader company strategy of thinking more holistically about how patients experience healthcare – not just how they experience a medicine – to advance neuroscience,” commented Steven Hildemann, Executive Vice President and Chief Medical Officer, Ipsen.

Committed to improving health outcomes

WCNR 2022 also marks the 10th anniversary of Ipsen’s Ixcellence Network®, an educational programme raising the standards of care for patients living with debilitating neurological disorders.

Since its launch, Ipsen has established 10 Ixcellence expert centres in Europe and Latin America, with nearly 800 physicians benefitting from the programme so far and improving the standard of care for thousands of patients with spasticity from around the world with its best-in-class injector training. 

Hamzah Baig, Vice President, Global Medical Affairs, Neuroscience, Ipsen, said: “With comprehensive and wide-ranging cutting-edge content, including use of advanced guidance techniques and implementation of multi-disciplinary care, the Ixcellence Network® is the only programme of its kind to have articles published in peer-reviewed journals; it is the gold-standard for advanced training in treating cervical dystonia and focal spasticity.”

As part of their commitment to physician education, Ipsen delivered an interactive educational training session at the WCNR, tailored around the management of complex patient cases.

Lecanemab: Clarity AD clinical study results

Results from Eisai’s large global Phase 3 confirmatory Clarity AD clinical study of lecanemab, an investigational anti-amyloid beta (Aβ) protofibril antibody for the treatment of mild cognitive impairment (MCI) due to Alzheimer’s disease (AD) and mild AD (collectively known as early AD) with confirmed presence of amyloid pathology in the brain, were presented at the 2022 Clinical Trials on Alzheimer’s Disease (CTAD) conference, in San Francisco, California. Simultaneously, the results were published in the New England Journal of Medicine read the full article here.

You can also read an interesting editorial in The Lancet: Lecanemab for Alzheimer’s disease: tempering hype and hope – ScienceDirect

Design of Clarity AD Study1

Eisai’s Clarity AD was a global confirmatory Phase 3 placebo-controlled, double-blind, parallel-group, randomised study in 1,795 people with early AD (lecanemab group: 898 placebo group: 897) at 235 sites in North America, Europe, and Asia. The participants were randomised 1:1 to receive either placebo or lecanemab 10-mg/kg IV biweekly, and the randomisation was stratified according to clinical subgroup (MCI due to AD or mild AD), presence or absence of concomitant approved AD symptomatic medication at baseline (e.g., acetylcholinesterase inhibitors, memantine, or both), ApoE4 status and geographical region.

The primary endpoint was change from baseline at 18 months in the CDR-SB (Clinical Dementia Rating Sum of Boxes), the global cognitive and functional scale, and key secondary endpoints were the change from baseline at 18 months in amyloid Positron Emission Tomography (PET) using centiloids, AD Assessment Scale – Cognitive Subscale 14 (ADAS-Cog14), AD Composite Score (ADCOMS) and AD Cooperative Study-Activities of Daily Living (ADCS MCI-ADL). In addition, longitudinal changes in brain tau pathology as measured by tau PET (n=257) and cerebrospinal fluid (CSF) biomarkers of AD pathology (n=281) were evaluated in optional sub-studies.

Efficacy results of Clarity AD2

Mean change of CDR-SB from baseline at 18 months as the primary endpoint was 1.21 and 1.66 for lecanemab (n=859) and placebo (n=875) groups, respectively. Lecanemab treatment resulted in highly statistically significant results, reducing clinical decline on the global cognitive and functional scale, compared with placebo at 18 months by -0.45 (95% Confidence Interval (CI): -0.67, -0.23; P=0.00005), representing a 27% slowing of decline. Starting as early as six months (difference: -0.17 [95% CI: -0.29, -0.05]; P<0.01), and increasing in absolute difference over time across all time points every 3 months, the treatment showed highly statistically significant changes in CDR-SB from baseline compared to placebo (all P-values are less than 0.01).

All key secondary endpoints also showed highly statistically significant results compared with placebo (P<0.001). In the amyloid PET sub-study, treatment with lecanemab showed statistically significant reduction in amyloid plaque burden at all timepoints starting at 3 months. Mean change in centiloids at 18 months was -55.5 and 3.6 for lecanemab (n=354) and placebo (n=344) groups, respectively (mean difference: -59.1 [95%CI: -62.6, -55.6]; P<0.00001). Lecanemab slowed decline of cognitive function by 26% on ADAS-Cog14 at 18 months (lecanemab (n=854), placebo (n=872); mean difference: -1.44 [95%CI: -2.27, -0.61]; P=0.00065). In the ADCOMS assessment, lecanemab slowed disease progression by 24% at 18 months (lecanemab (n=857), placebo (n=875); mean difference: -0.050 [95% CI: -0.074, -0.027; P=0.00002]). Lecanemab slowed decline of activities of daily living by 37% on ADCS MCI-ADL at 18 months (lecanemab (n=783), placebo (n=796); mean difference: 2.016 [95%CI: 1.208, 2.823]; P<0.00001).

Safety Results of Clarity AD3

The most common adverse events (>10%) in the lecanemab group were infusion reactions (lecanemab (n=898): 26.4%; placebo (n=897): 7.4%), ARIA-H (combined cerebral microhaemorrhages, cerebral macrohaemorrhages, and superficial siderosis; lecanemab (n=898): 17.3%; placebo (n=897): 9.0%), ARIA-E (edema/effusion; lecanemab (n=898): 12.6%; placebo 9n=897): 1.7%), headache (lecanemab (n=898): 11.1%; placebo (n=897): 8.1%), and fall (lecanemab (n=898): 10.4%; placebo (n=897): 9.6%). Infusion reactions were largely mild-to-moderate (grade 1-2: 96%) and occurred on the first dose (75%).

During the study period, deaths occurred in 0.7% and 0.8% of participants in the lecanemab and placebo groups, respectively and no deaths were related to lecanemab or occurred with amyloid-related imaging abnormalities (ARIA) in 18-month double-blind study period. Serious adverse events were experienced by 14.0% of participants in the lecanemab group and 11.3% of participants in the placebo group. Treatment-emergent adverse events occurred in 88.9% and 81.9% of participants in the lecanemab and placebo groups, respectively. Treatment-emergent adverse events leading to drug withdrawal occurred in 6.9% and 2.9% of participants in the lecanemab and placebo groups, respectively.

Overall, lecanemab’s ARIA incidence profile was within expectations based on the Phase 2b results.

Eisai serves as the lead of lecanemab development and regulatory submissions globally with both Eisai and Biogen co-commercialising and co-promoting the product and Eisai having final decision-making authority.

The results of Clarity AD are very encouraging, representing the first time a treatment has modified Alzheimer’s disease. The news is great for not only patients but those in the research field as well.

Prof. John Hardy, Chair of Molecular Biology of Neurological Disease, UCL

References

  1. Irizarry M. Clarity AD: Clinical Trial Background and Study Design Overview. Presented at: 15th Annual Clinical Trials on Alzheimer’s Disease (CTAD) Conference, November 29 – December 2, 2022, San Francisco, California, USA.
  2. van Dyck C. Lecanemab for the Treatment of Early Alzheimer’s Disease: Topline Efficacy Results from Clarity AD. Presented at: 15th Annual Clinical Trials on Alzheimer’s Disease (CTAD) Conference, November 29 – December 2, 2022, San Francisco, California, USA.
  3. Sabbagh M. Safety Profile of Lecanemab in Early Alzheimer’s Disease. Presented at: 15th Annual Clinical Trials on Alzheimer’s Disease (CTAD) Conference, November 29 – December 2, 2022, San Francisco, California, USA.

Zilucoplan for the treatment of generalised myasthenia gravis in adult patients

FDA acceptance of new drug application and EMA MAA validation

  • New drug application (NDA) for zilucoplan seeks approval for the treatment of generalised myasthenia gravis (gMG) in adult patients who are acetylcholine receptor antibody positive (AChR-Ab+) 
  • Acceptance by U.S. Food and Drug Administration (FDA) follows the recent European Medicines Agency (EMA) validation of Marketing Authorization Application (MAA) for treatment of adult patients with AChR-Ab+ gMG and who require treatment in addition to steroids or non-steroidal immunosuppressants 
  • Both NDA and MAA are based on pivotal Phase 3 RAISE study in gMG, which demonstrated treatment with zilucoplan resulted in clinically meaningful and statistically significant improvements in key MG-specific outcomes compared to placebo
  • UCB expects to receive feedback from the agencies in Q4 of 2023 

On 14th November 2022, UCB announced that the U.S. Food and Drug Administration (FDA) has accepted for review the New Drug Application (NDA) for its investigational treatment, zilucoplan. 

Zilucoplan is a subcutaneous (SC), self-administered peptide inhibitor of complement component 5 (C5 inhibitor) for the treatment of adult patients with acetylcholine receptor antibody positive (AChR-Ab+) generalised myasthenia gravis (gMG). 

In 2019, the U.S. FDA granted orphan drug designation to zilucoplan for the treatment of MG.1 The safety and efficacy of zilucoplan have not been established and it is not currently approved for use in any indication by any regulatory authority worldwide.

This acceptance follows the recent European Medicines Agency (EMA) validation of the Marketing Authorization Application (MAA) for zilucoplan for the treatment of adult patients with AChR-Ab+ gMG and who require treatment in addition to steroids or non-steroidal immunosuppressants. Validation confirms that the application is complete and the formal review process by the EMA’s Committee for Medicinal Products for Human Use (CHMP) can begin. Orphan designation was granted in 2022 by the European Commission to zilucoplan for the treatment of myasthenia gravis.2

gMG is a chronic and unpredictable auto-immune disease in which pathogenic autoantibodies can impair synaptic transmission at the neuromuscular junction by targeting specific proteins on the post-synaptic membrane. This disrupts the ability of the nerves to stimulate the muscle and results in a weaker contraction.4 People living with MG can experience a variety of symptoms, including drooping eyelids, double vision, and difficulty in swallowing, chewing and talking, as well as severe muscle weakness that can result in life-threatening weakness of the muscles of respiration.3 In the U.S. the prevalence of MG is estimated at 14 to 20 per 100,000 population; approximately 36,000 to 60,000 cases.4 In Europe, the prevalence is estimated at 10 per 100,000 population.5

People living with gMG experience high treatment burden, on top of the debilitating impact of the condition, and there is a clear need for additional targeted treatments to support the gMG community. Our goal is to provide a solution that can help meet these needs and transform lives. The acceptance of the NDA by the FDA as well as the acceptance of the MAA by the EMA, brings us one step further on our journey towards approval for this medicine. We look forward to working with the FDA and EMA to help bring this important new treatment option to patients.”

Charl van Zyl, Executive Vice President Neurology Solutions & Head of EU/International Markets, UCB.

The NDA and MAA are based on data from the pivotal Phase 3 RAISE study (NCT04115293), which demonstrated at week 12 that treatment with zilucoplan (0.3 mg/kg daily) resulted in clinically meaningful and statistically significant improvements in key gMG-specific outcomes compared with placebo in patients with AChR-Ab+ gMG. The study met its primary endpoint with zilucoplan showing a placebo-corrected mean improvement of 2.09 points in the Myasthenia Gravis Activities of Daily Living (MG-ADL) score at week 12 (p<0.001).6

Zilucoplan demonstrated a favorable safety and tolerability profile, showing a similar rate of treatment-emergent adverse events (TEAEs) between zilucoplan (76.7%) and placebo (70.5%). The most common TEAEs were injection site bruising, headache, and diarrhea. Rates of treatment discontinuation due to a TEAE were low and all patients who completed the 12-week treatment period have entered the ongoing RAISE-XT open-label extension study (NCT04225871).6,7

In the RAISE study, 174 adult patients were randomised to receive daily SC, self-administered doses of placebo (N=88) or zilucoplan 0.3 mg/kg (N=86). Patient demographics and baseline disease characteristics were generally balanced between treatment arms.6

As a complement C5 inhibitor, zilucoplan is a targeted therapy that inhibits key components in the underlying pathophysiology of gMG, addressing the underlying mechanism of neuromuscular junction damage.8,9

UCB anticipates making regulatory filings for zilucoplan in gMG in Great Britain, Japan, and rest of the world from Q3 2022 onwards. 

Alongside zilucoplan, UCB is also investigating rozanolixizumab, a SC-administered, humanized monoclonal antibody that specifically binds, with high affinity, to human neonatal Fc receptor (FcRn), as a potential treatment for gMG. UCB anticipates filing regulatory submissions for rozanolixizumab later this year. 

About the zilucoplan RAISE study 6,7

The RAISE study (NCT04115293) is a multi-center, Phase 3, randomized, double-blind, placebo-controlled study to confirm the efficacy, safety, and tolerability of zilucoplan in patients with AChR-Ab+ gMG. Patients were randomized in a 1:1 ratio to receive daily subcutaneous (SC) doses of 0.3 mg/kg zilucoplan or placebo for 12 weeks.

The primary endpoint for the RAISE study is change from baseline to Week 12 in the Myasthenia Gravis-Activities of Daily Living (MG-ADL) score. Secondary endpoints include change in the Quantitative Myasthenia Gravis (QMG) score, the Myasthenia Gravis Composite (MGC) and the Myasthenia Gravis Quality of Life 15 revised (MG-QoL15r) score from baseline to Week 12,  time to rescue therapy, the proportion with minimal symptom expression (MSE) (defined as MG-ADL of 0 or 1), the proportion with a ≥3-point reduction in MG-ADL and the proportion with a ≥5-point reduction in QMG without rescue therapy, all measured at Week 12. The secondary safety endpoint is incidence of TEAEs. Patients who completed the 12 week RAISE trial had the possibility to enter the open label extension study, RAISE-XT. 

For more information about the trial visit https://clinicaltrials.gov/ct2/show/NCT04115293


References:

  1. US Food and Drug Administration https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=699319. Accessed October 2022
  2. Data on file.
  3. Hansen JS, et al. Mortality in myasthenia gravis: A nationwide population-based follow-up study in Denmark. Muscle Nerve. 2016;53:73-77.
  4. Myasthenia Gravis Foundation of America. Clinical Overview of MG. https://myasthenia.org/Professionals/Clinical-Overview-of-MG. Accessed August 2022 
  5. Salari N, et al. Global prevalence of myasthenia gravis and the effectiveness of common drugs in its treatment: a systematic review and meta-analysis. J Transl Med 19, 516 (2021). https://doi.org/10.1186/s12967-021-03185-7. Accessed September 2022.
  6. Vu T, et al. Efficacy and safety of zilucoplan in myasthenia gravis: Responder analysis from the randomized Phase 3 RAISE trial. Poster 200, AANEM 2022.
  7. Weiss MD, et al, Quality of life outcomes in RAISE: A double-blind randomized, placebo-controlled study of zilucoplan in gMG. Oral presentation. MGFA Scientific Session, AANEM 2022.
  8. Tannemaat et al. Emerging therapies for autoimmune myasthenia gravis: Towards treatment without corticosteroids. Neuromuscul Disord. 2020;30(2):111-119
  9. Howard J, et al. Clinical Effects of the Self-administered Subcutaneous Complement Inhibitor Zilucoplan in Patients With Moderate to Severe Generalized Myasthenia Gravis: Results of a Phase 2 Randomized, Double-Blind, Placebo-Controlled, Multicenter Clinical Trial. JAMA Neurol 2022 1;77(5)
  10. ClinicalTrials.gov. 2022. A Study to Test Efficacy and Safety of Rozanolixizumab in Adult Patients With Generalized Myasthenia Gravis. https://clinicaltrials.gov/ct2/show/NCT03971422. Accesed October 2022 
  11. Chen J, et al. Incidence, mortality, and economic burden of myasthenia gravis in China: A nationwide population-based study. Lancet Reg Health West Pac: 2020;5:100063.
  12. National institute of Neurological Disorders and Stroke. 2022. Myasthenia Gravis Fact Sheet. https://www.ninds.nih.gov/myasthenia-gravis-fact-sheet. Accessed October 2022.
  13. Dong D, et al. Gender differences in quality of life among patients with myasthenia gravis in China. Health and Quality of Life Outcomes 2020 18;296
  14. Myasthenia Gravis Foundation of America. MG Quick Facts. https://myasthenia.org/MG-Education/MG-Quick-Facts Accessed October 2022
  15. Smith B, et al. Generation and characterization of a high affinity anti-human FcRn antibody, rozanolixizumab, and the effects of different molecular formats on the reduction of plasma IgG concentration. MAbs. 2018;10:1111-30.
  16. Kiessling P, et al. The FcRn inhibitor rozanolixizumab reduces human serum IgG concentration: A randomized phase 1 study. Sci Transl Med. 2017;9(414:eaan1208).
  17. US Food and Drug Administration https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=669918. Accessed August 2022
  18. European Medicines Agency, EU/3/20/2272: Orphan designation for the treatment of myasthenia gravis https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu3202272. Accessed August 2022 

Upstaza™ gene therapy granted marketing authorisation for AADC deficiency by MHRA

  • First gene therapy infused directly into the brain for the treatment of AADC deficiency
  • MHRA authorisation follows recent European Union authorisation
  • AADC deficiency is a rare, fatal disorder that arrests motor development causing severe disability

PTC Therapeutics, Inc. announced on 17th November that Upstaza™ (eladocagene exuparvovec) was granted authorisation by the Medicines and Healthcare Products Regulatory Agency (MHRA) in Great Britain. Upstaza is the first and only approved disease-modifying treatment for aromatic L-amino acid decarboxylase (AADC) deficiency and the first marketed gene therapy directly infused into the brain. It is approved for patients 18 months and older.

“We are thrilled with the MHRA’s rapid authorisation of Upstaza,” said Stuart W. Peltz, Ph.D., Chief Executive Officer, PTC Therapeutics. “Patients in the UK with AADC deficiency are one step closer to having access to a much-needed disease modifying therapy. This is another milestone towards our commitment to advance innovative treatments and improve outcomes for people living with rare diseases.”

During Upstaza clinical studies, patients went from not achieving any developmental motor milestones to demonstrating a mastery of clinically meaningful motor skills, including the ability to ambulate independently. Milestone achievements including cognitive and language acquisition occurred from as early as three months following treatment, with clinically significant improvements shown to continue up to ten years after treatment. Upstaza also reduced symptoms that cause potentially life-threatening and morbid complications.

“The approval of an AADC deficiency gene therapy in the UK will provide the opportunity to transform the prognosis for those born and living with this disease, and we are hopeful for access in the coming months. Without treatment, most children born with AADC deficiency will have difficulty with their development and many of the symptoms can be distressing and life-threatening. The impact on those living with AADC and their communities is significant, with children facing frequent hospitalisations, emergency visits and requiring a multi-disciplinary team of highly trained specialists,” stated Kirsty Hoyle, CEO, Metabolic Support UK

Upstaza has marketing authorisation in all 27 European Union member states, as well as Iceland, Norway, Liechtenstein, and Northern Ireland.  

Patients in the UK with AADC deficiency are one step closer to having access to a much-needed disease modifying therapy. This is another milestone towards our commitment to advance innovative treatments and improve outcomes for people living with rare diseases.

Stuart W. Peltz, Ph.D., Chief Executive Officer, PTC Therapeutics.

References:

  1. Tai CH, et al. Long-term efficacy and safety of eladocagene exuparvovec in patients with AADC deficiency. Mol Ther. 2022;30(2):509-518.
  2. Chien et al. AGIL-AADC gene therapy results in sustained improvements in motor and developmental milestones through 5 years in children with AADC deficiency. Poster presented at the 48th Annual Meeting of the Child Neurology Society, Charlotte, NC, USA, Oct 23-26, 2019.
  3. Chien YH, et al. Efficacy and safety of AAV2 gene therapy in children with aromatic L-amino acid decarboxylase deficiency: an open-label, phase 1/2 trial. Lancet

About Upstaza™ (eladocagene exuparvovec)

Upstaza is a one-time gene replacement therapy indicated for the treatment of patients aged 18 months and older with a clinical, molecular, and genetically confirmed diagnosis of aromatic L‑amino acid decarboxylase (AADC) deficiency with a severe phenotype. It is a recombinant adeno-associated virus serotype 2 (AAV2)-based gene therapy, containing the human DDC gene.1 It is designed to correct the underlying genetic defect, by delivering a functioning DDC gene directly into the putamen, increasing the AADC enzyme and restoring dopamine production.2,3

The efficacy and safety profile of Upstaza has been demonstrated across clinical trials and compassionate use programmes.1 The first patient was dosed in 2010. In clinical trials, Upstaza demonstrated transformational neurological improvements. The most common side effects were initial insomnia, irritability and dyskinesia.

Administration of Upstaza occurs through a stereotactic surgical procedure, a minimally invasive neurosurgical procedure used for the treatment of a number of paediatric and adult neurological disorders. The Upstaza administration procedure is performed by a qualified neurosurgeon in centers specialised in stereotactic neurosurgery.

About aromatic L-amino acid decarboxylase (AADC) deficiency

AADC deficiency is a fatal, rare genetic disorder that typically causes severe disability and suffering from the first months of life, affecting every aspect of life – physical, mental and behavioural. The suffering of children with AADC deficiency may be exacerbated by: episodes of distressing seizure-like oculogyric crises causing the eyes to roll up in the head, frequent vomiting, behavioural problems, and difficulty sleeping.

The lives of affected children are severely impacted and shortened. Ongoing physical, occupational and speech therapy, and interventions, including surgery, also are often required to manage potentially life-threatening complications such as infections, severe feeding and breathing problems.

About PTC Therapeutics, Inc.

PTC is a science-driven, global biopharmaceutical company focused on the discovery, development and commercialization of clinically differentiated medicines that provide benefits to patients with rare disorders. PTC’s ability to innovate to identify new therapies and to globally commercialise products is the foundation that drives investment in a robust and diversified pipeline of transformative medicines. PTC’s mission is to provide access to best-in-class treatments for patients who have little to no treatment options.

First autopsy-confirmed results for identifying Alzheimer’s in presence of other co-morbid pathologies using DISCERN test

SYNAPS Dx (SDx), a privately held company focused on the research, development and commercialisation of a diagnostic test for Alzheimer’s disease (AD), is pleased to announce the publication of “Morphometric imaging biomarker identifies Alzheimer’s disease even among mixed dementia patients” in Scientific Reports.

Up until now, there have been no autopsy-validated tests shown to accurately identify AD in the presence of mixed dementia, or dementia from one or more co-existing causes, affecting over half of patients diagnosed with AD. We are pleased to share this groundbreaking data showing that the MI biomarker demonstrated specificity and sensitivity of 100% (p<.001) when compared to the NIH Gold Standard criteria for AD in distinguishing AD from non-AD patients, even in the presence of other co-morbid pathologies.

Dr. Daniel Alkon, chief scientific advisor, SDx and corresponding author of the paper.

Dr. Alkon further explains that DISCERN™ is a CLIA-certified diagnostic skin test comprised of three assays that assess several factors related to synaptic and neuronal loss that impact cognitive loss, as well as regulators of amyloid plaque and neurofibrillary tangles in the brain. The test is autopsy-validated to inform a definitive diagnosis of AD, even in people recently diagnosed with dementia. The DISCERN AD biomarkers also depend on factors that regulate amyloid plaque and tau formation, hallmarks of AD at autopsy.

“DISCERN is an important breakthrough for the AD community because there are currently no other autopsy-validated tests that are highly sensitive and highly specific for identifying AD in people recently diagnosed with dementia or mixed-dementia,” says Frank Amato, CEO and president, SDx. “With expected FDA-approved treatments for AD on the horizon, in addition to determining the presence of amyloid plaques consistent with AD, patients will likely need an accurate AD diagnosis to determine if amyloid targeted drugs are appropriate for them.”

Amato explains that currently, most patients with dementia are managed in primary care where due to the lack of availability of diagnostic tools, more than half are misdiagnosed.

“While blood biomarkers have the potential to help clinicians screen for the presence of amyloid plaque or tau, hallmarks of AD, they do not diagnose mild cognitive impairment (MCI) or early AD dementia with high accuracy,” Dr. Alkon continues. “Well-defined cohorts such as the oldest old individuals can be entirely cognitively normal – i.e. not at all demented – but have abundant amyloid plaques and tau tangles at autopsy.”

“With use of the DISCERN test, which relates to loss of synaptic networks as well as amyloid and tau, primary care clinicians have indicated a greater confidence in the diagnosis of AD, potentially avoiding unnecessary, invasive and expensive diagnostic procedures and identifying patients with AD dementia who are appropriate candidates for treatment,” says Dr. Alkon.

About SYNAPS Dx
SYNAPS Dx is a privately held company focused on the research, development and commercialisation of a diagnostic test for Alzheimer’s disease (AD). The Company offers DISCERN™, the first highly accurate, minimally invasive test supporting a clinician’s definitive diagnosis of AD versus other forms of dementia, even in people recently diagnosed with dementia. SYNAPS Dx’s laboratory is certified under the Clinical Laboratory Improvement Amendments (CLIA) as qualified to perform high complexity clinical laboratory testing. Physicians and patients seeking more information can visit https://discerntest.com/. For more information on the Company, visit https://www.synapsdx.com/. Contact: info@synapsdx.com.

Tecfidera lowers risk of first MS symptoms in RIS

Treatment with Tecfidera (dimethyl fumarate) significantly reduces the risk of experiencing the first multiple sclerosis (MS) symptoms in adults with radiologically isolated syndrome (RIS), according to data from a Phase 4 clinical trial which may lead to changes in practice.

RIS is a condition in which patients have MS-like lesions on MRI scans, but lack typical symptoms of the disease.

This is the first clinical trial demonstrating the benefit of disease-modifying therapies (DMTs) in people with RIS — the “earliest detectable pre-clinical phase of multiple sclerosis,” Darin Okuda, MD, the trial’s principal investigator, said in an oral presentation at the 38th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS).

Okuda is a Professor of Neurology and the Director of the MS and Neuroimmunology Imaging Programme, as well as Director of the Neuroinnovation Programme, at University of Texas Southwestern Medical Center, in Dallas, US.

The trial’s results, “Multi-center, randomized, double-blinded assessment of dimethyl fumarate in extending the time to a first clinical demyelinating event in radiologically isolated syndrome (ARISE),” were shared by Okuda at the ECTRIMS 2022 Congress, held October 26–28, both virtually and in Amsterdam, Netherlands. Biogen, which developed and markets Tecfidera as a DMT, funded the trial.

EMD Serono highlights new data for Evobrutinib

First BTKi to demonstrate sustained clinical benefit for people with RMS through three and a half years of treatment

  • Phase II clinical trial data of evobrutinib demonstrated low disease activity and stable EDSS, with NfL levels, a marker of neuronal injury, remaining low in people with RMS after three and a half years of therapy
  • Late-breaking data showed evobrutinib-treated patients mounted an antibody response to mRNA COVID vaccinations similar to that of healthy subjects
  • Evobrutinib is an investigational highly-selective, oral, CNS-penetrant BTK inhibitor with the potential to become a best-in-class treatment option for people living with RMS

EMD Serono, the Healthcare business sector of Merck KGaA, Darmstadt, Germany, in the US and Canada, announced on 26th October 2022 findings which demonstrated that annualised relapse rates (ARR) remained low and Expanded Disability Status Scale (EDSS) scores were stable in people with relapsing multiple sclerosis (RMS) treated with investigational evobrutinib through more than three and half years. Additionally, the number of T1 gadolinium-enhancing (Gd+) lesions and T2 lesion volume remained low for the duration of the open-label extension (OLE) of the Phase II clinical trial. These data, presented at the 38th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS), suggest the long-term positive benefits of evobrutinib for people with RMS as a potential best-in-class therapy.

Evobrutinib is an oral, highly selective, central nervous system (CNS) penetrant immunomodulator that has the potential to become a safe and highly efficacious treatment option for RMS by addressing both peripheral and central drivers of inflammation through inhibition of Bruton’s tyrosine kinase (BTK) signaling in B cells as well as microglia. The dual-faceted approach of evobrutinib may offer better control of silent progression of the disease in between attacks on top of strong relapse control in people living with RMS.

Disease progression is a top concern in the MS community. Learning more about silent disease progression without relapses will help us further our understanding of MS, along with potential treatments, as it has not just physical but also cognitive and mental deleterious impact. In this longest-running and most extensive analysis of any BTK inhibitor in development for RMS, evobrutinib maintained disease stability for up to three and half years. It also has the potential to directly address smoldering inflammation in RMS which contributes to the silent causes of disease progression. It has previously shown promising results in targeting central inflammation, including through its modulatory effects on microglia.

Patrick Vermersch, MD, PhD, Vice President, Research in Biology and Health, University of Lille

The OLE of the Phase II clinical trial evaluated the long-term treatment effect of evobrutinib on ARR, EDSS scores and several magnetic resonance imaging (MRI) outcomes, in people with RMS:

  • Patients assigned to the initial 75mg twice-daily arm, maintained a low ARR of 0.13 throughout the course of the OLE. In addition, switching from 75mg once-daily to 75mg twice-daily in the OLE reduced ARR from 0.19 to 0.09
  • Overall, mean EDSS scores, as well as MRI lesion activity remained low and stable throughout the entire study

These data points further strengthen the observations made previously that maximal BTK occupancy throughout the dosing interval achieved with twice-daily dosing is correlated to higher ARR reductions with evobrutinib.

The Company also presented new long-term data from the Phase II clinical trial OLE which found reductions of blood neurofilament light chain (NfL) levels, a key biomarker which may predict future brain volume loss and disease progression. Patients experienced sustained and ongoing reductions in blood NfL levels compared to the double-blind period (DBP) and OLE baseline values. A dose of 75mg twice-daily significantly reduced NfL levels from week 12 (DBP), compared to placebo/evobrutinib 25mg once-daily showing an early dose-response. This reduction in NfL provides evidence evobrutinib may reduce neuronal damage in people with RMS.

“This is the first time that evidence of sustained efficacy out to three and a half years could be shown with a BTK inhibitor in RMS,” said Jan Klatt, Senior Vice President, Head of Development Unit Neurology and Immunology, Merck KGaA, Darmstadt, Germany. “Combined with our previous data demonstrating reduced volume of slowly expanding lesions, indicative of an effect on microglia, and reduced neurofilament levels, a marker of neuronal injury, we are confident evobrutinib has the potential to offer best-in-class efficacy for people living with RMS.”

Late-breaking data from a post hoc analysis of vaccinated patients (n=24) in the Phase II OLE were also presented, showing 96% of people with RMS treated with evobrutinib (75mg twice daily) were able to mount an antibody response following two doses of an mRNA COVID-19 vaccine, similar to untreated RMS patients and healthy subjects. The increase in antibody response in seronegative and seropositive patients demonstrated a preserved response to novel and recall antigens. This is the first time this could be shown with a BTK inhibitor in RMS and these findings are consistent with the modulation of B cell function, providing a potentially alternative treatment to B cell depletion approaches.


About Evobrutinib
Evobrutinib is an oral, highly selective CNS-penetrant inhibitor of Bruton’s tyrosine kinase (BTK) in clinical development as a potential treatment for relapsing multiple sclerosis (RMS). It is the first BTK inhibitor to demonstrate clinical efficacy in the largest Phase II study with follow-up beyond three years as well as demonstrate an impact on early biomarkers of ongoing central inflammation that correlate with disease progression. Evobrutinib is designed to modulate B cell responses such as proliferation and antibody and cytokine release, as well as modulate macrophage/microglia activation. It significantly reduced SEL volume and levels of blood NfL, markers of ongoing central inflammation and neurodegeneration that predict long-term disability. Evobrutinib was optimized for efficacy through the most comprehensive BTK inhibitor dose-finding study in RMS which demonstrated 75mg twice-daily dosing achieves maximal efficacy across endpoints by sustaining BTK inhibition throughout the dosing interval (>95% BTK occupancy maintained in 98% of patients before next dose). It is currently under clinical investigation and is not approved for any use anywhere in the world.

About the Open-Label Extension (OLE) Phase II Clinical Trial
In the 48 week double-blind period (DBP), patients with RMS were assigned to one of five treatment groups: placebo (switching to 25mg once-daily evobrutinib after 24 weeks), 25mg or 75mg once-daily evobrutinib, 75mg twice-daily evobrutinib, or open-label dimethyl fumarate (120mg twice daily for the first week and 240mg twice daily thereafter). At week 48, patients could enter the OLE and received evobrutinib 75mg once daily for a mean time of 49.8 weeks before switching to 75mg twice daily for the remainder of the OLE.

Laser Therapy for Epilepsy now available on the NHS

NHS England has accepted the recommendation of the Clinical Priorities Advisory Group to roll out LITT (laser interstitial thermal therapy) on the NHS. LITT is a laser treatment for epilepsy offering a less invasive alternative to conventional neurosurgery. It is a stereotactic procedure with the potential for seizure reduction results similar to those of open resection, with fewer side-effects or complications.

LITT allows for individualised neurological treatment with a small wound, a much faster recovery time, less pain, and minimal risk of infection, adverse neurocognitive deficits, or other side effects.

This pioneering laser beam treatment for epilepsy patients is life-changing and will offer hope to hundreds of people every year who have not had success in preventing seizures with traditional drugs. By replacing invasive neurosurgery with a cutting-edge laser therapy, allowing clinicians to better target the parts of the brain causing the epilepsy, we not only dramatically reduce risks to these patients, but drastically reduce their recovery time both in and out of hospital.

The treatment is yet another example of how the NHS continues to deliver on its NHS Long Term Plan commitment to secure the latest medical innovations for patients while also using our commercial means to ensure value for money.

Professor Sir Stephen Powis, NHS national medical director

Find the clinical commissioning policy here.

AssistiveTech in the UK

Ecosystem Overview and ATLAS Advocacy Launched in House of Lords

AssistiveTech poised for rapid growth in the UK, tackling key socio-economic challenges.

The official launch event and key findings of the new ‘AssistiveTech in the UK’ interactive report, developed by ATLAS (Assistive Technology, Longevity and Ageing Society) with the support of Aging Analytics Agency and Deep Knowledge Philanthropy was launched in the House of Lords on Tuesday October 4th, 2022.

The event brought together key industry and ecosystem participants, thought leaders, founders of AssistiveTech and AgeTech organisations across governance, policy, charity, academia and industry in a bid to harness the power of technology for social good.

Co-launched with the report was UK-based advocacy initiative, ATLAS, which seeks to help inform and guide the public and private sectors to embrace the UK’s growing Assistive tech, Longevity and Agetech industries.

TLAS’ analysis depicts five segments of companies within AssistiveTech in the UK: Assistive Care Services, Devices and Apps, Education and Consulting, Tech-Enabled Home Care, and Smart Homes technologies.

The report provides insights on numerous AssistiveTech ecosystem participants and stakeholders, major trends and obstacles, and highlights the government’s developmental role in this emerging ecosystem.