Thursday, July 25, 2024

Ashya King update

In 2014, the parents of five-year-old brain tumour patient Ashya King removed him from NHS care and travel abroad instead.

Ashya had successfully undergone surgery to remove the tumour but the hospital planned to treat him with radiotherapy to ensure the cancer did not return.

Naghmeh and Brett King wanted their son to get proton beam therapy instead, a similar treatment to radiotherapy which is thought to have fewer side effects, but was not offered on the NHS at the time.

Ashya was taken out of Southampton General Hospital and the family travelled to France. This sparked an international manhunt which resulted in the parents being arrested in Spain.

But the High Court eventually ruled that Ashya could get proton therapy in the Czech Republic. A decade later, he is still alive and the NHS now offers some brain cancer patients proton therapy.

https://www.dailymail.co.uk/health/article-13435169/truth-foreign-cancer-clinics-terminally-ill-child-experts-alarm-unproven-treatments.html

See: https://childnervoussystem.blogspot.com/2024/07/sounding-alarm-about-unorthodox-and.html
https://childnervoussystem.blogspot.com/2020/01/ashya-kings-3-year-remission.html
https://childnervoussystem.blogspot.com/2017/07/ashya-king-and-freya-bevan-updates.html
https://childnervoussystem.blogspot.com/2015/02/ashya-king-redux_24.html

Sounding the alarm about "unorthodox" and "unproven" treatments for terminally ill children

Desperate families are raising millions of pounds every year to send their terminally-ill children abroad for 'unproven' brain cancer treatments which 'do not work', experts warn.

Overseas clinics are offering places on clinical trials for experimental drugs to young patients – but making them pay for every aspect of the care.

All have been told by NHS doctors that there is nothing more that can be done to slow the disease – so feel they have little other choice.

The medical centres – often in Mexico, Germany and the US – charge a high price, with families stumping up as much as £1 million. Most raise the cash via crowdfunding websites such as GoFundMe and JustGiving.

However, top brain tumour experts claim many of these clinics offer 'unorthodox' medicines and surgical procedures and sometimes 'take advantage' of families looking for a shred of hope.

In some cases, these treatments have a high rate of side effects, hospitalising some children, they say. And in many cases these therapies do not extend lives.

Some child brain tumour doctors told The Mail on Sunday they know of families who sought help from clinics which offered ineffective low-carbohydrate diets and cannabis oil treatments.

According to GoFundMe, around £500,000 is raised every year to fund private treatment and associated costs for children with one form of rare brain tumour alone, diffuse intrinsic pontine glioma (DIPG).

Our findings come after the Government announced last week it would spend £40 million on brain tumour research in an effort to improve the poor outcomes for the disease.

Brain Tumour Research is calling on the Government to use the money to fund more clinical trials for children with the disease, so parents are not forced to raise these large sums – or placed in extreme financial difficulty.

Childhood brain tumours are some of the trickiest diseases in the world to treat – but one type is arguably even worse than the rest.

Every year, around 30 British children are diagnosed with a diffuse midline glioma, a fast-growing and incurable brain tumour, also known as a DIPG.

While it can affect adults, the disease is most common in young children – for reasons still not understood.

The first signs are often headaches, seizures, sudden mental or behavioural changes, as well as vision or speech problems.

These tumours commonly grow around a crucial part of the brain stem known as the pons, which links the brain to the spinal cord.

Due to this sensitive position, most surgeons will not operate on DIPG patients.

The NHS typically only offers radiotherapy in such cases, which can slow the disease, but cannot cure it. Even with radiotherapy, most DIPG patients live no longer than 18 months after diagnosis.

'We know of at least five families who last year were raising money to travel overseas. The numbers taking these last-chance options shames the UK,' says Dr Karen Noble, Director of Research at Brain Tumour Research.

'Of course parents will travel the four corners of the Earth to find hope, but this hope must be based on science and clinical evidence. Our position isn't one of criticism of the families, it is one of complete understanding and outrage that this situation has been reached as the UK slides down the list of countries where clinical trials are being instigated.'

'Sadly the majority of overseas treatments do not extend the lives of children any more than standard NHS treatment,' says Dr Fernando Carceller, consultant paediatric neuro-oncologist at The Royal Marsden NHS Foundation Trust.

'And there are cases where children experience uncomfortable side effects while taking these drugs.'

Every year, around 16,000 people in the UK are diagnosed with a brain tumour. Roughly 400 of these are in children.

Fewer than a fifth of patients survive longer than five years after diagnosis.

Brain tumours are also the biggest cancer killer of children.

Clinical trials – which enable patients to access new, experimental treatments before they are approved – can offer some hope.

However there has been a 40 per cent reduction in new trials since 2017, due to a lack of funding and the impact of the Covid pandemic. News about experimental therapies often spreads through Facebook groups dedicated to brain tumour families.

Prior to the pandemic, experts say there was a surge in demand for an unproven procedure in Mexico which involved injecting chemotherapy drugs directly into the arteries that carry blood to the brain.

This was based on the theory that many cancer drugs are ineffective because they cannot pass the blood-brain barrier – a membrane filter which blocks unwanted substances from reaching the brain.

In one case, the British family of nine-year-old Lucy Moroney raised £300,000 in 2017 to send her to Mexico for the procedure.

However, less than a year after starting the treatment, Lucy died. There is still no high-quality evidence that the procedure benefited patients.

Experts say that, since then, there have been other new treatment trends that have led to parents travelling abroad.

'I've had parents asking about ketogenic [low-carbohydrate] diet and CBD [cannabis] oil clinics for their children,' says Dr Carceller.

'These trends come and go but families will cling on to anything. They don't want their children to miss out on an experimental treatment which could save them.'

He added: 'The majority of physicians at these clinics would not take on children who are too unwell to benefit. But there are doctors out there taking advantage of the desperate situation these families are in.'

Unlike when going abroad, taking part in clinical trials in the UK does not cost anything.

Eight-year-old Rudi Abbot from Edinburgh was told by the NHS in 2022 that his brain cancer could not be cured, and was sent to Seattle in the US for an experimental immune-boosting drug trial after his parents raised £110,000.

Rudi remained on the medicine for just four months before doctors concluded it had no effect. He died less than a year later.

While experts sympathise with parents doing what they can to save their children, some question whether these crowdfunding ventures are being put to good use.

'These are huge lumps of money we're talking about,' says Dr Carceller. 'In some cases, it would be enough to open a new trial in the UK in itself.

There is also a risk the patient could fall severely unwell while abroad – placing the family in even greater financial difficulty.

In 2022, George Fox, 13, from Bedfordshire travelled to Los Angeles after his parents raised £300,000 on GoFundMe to get him on an unapproved cancer therapy.

However, during the journey, his condition deteriorated and he was taken into emergency care. He never began the treatment. The family were left with a £500,000 medical bill, paid by raising another £200,000 through crowdfunding. He died months later.

'Families are away from their support network and do not have the NHS if a child deteriorates overseas,' says Prof Darren Hargrave, a paediatric neuro-oncologist at Great Ormond Street in London. 'They may find they face unexpected and massive additional costs and stress.'

However, there are some children who do benefit from travelling abroad for treatment.

In 2014, the parents of five-year-old brain tumour patient Ashya King removed him from NHS care and travel abroad instead.

Ashya had successfully undergone surgery to remove the tumour but the hospital planned to treat him with radiotherapy to ensure the cancer did not return.

Naghmeh and Brett King wanted their son to get proton beam therapy instead, a similar treatment to radiotherapy which is thought to have fewer side effects, but was not offered on the NHS at the time.

Ashya was taken out of Southampton General Hospital and the family travelled to France. This sparked an international manhunt which resulted in the parents being arrested in Spain.

But the High Court eventually ruled that Ashya could get proton therapy in the Czech Republic. A decade later, he is still alive and the NHS now offers some brain cancer patients proton therapy.

Experts say these cases are rare and that the blunt truth is that, in many cases, there is nothing that can be done for patients – particularly those with DIPG tumours.

'When I see children with these tumours, I want to make sure the child is cared for,' says Prof Hargrave. 'We want to make sure they don't suffer and that the family is supported.

'We might talk about experimental trials as an option, but we'd be honest about the chances of trial therapies working.'

Experts agree though that it is crucial the Government makes more funding available for brain cancer clinical trials in the UK.

'Trials are not a panacea but they are the best solution we have,' says Dr Carceller.

'The more patients we can get on trials in the UK, the greater chance we have at finding new cures.'

The first sign that seven-year-old Jasmine Freeman was anything but perfectly healthy was when she began to experience double vision in February 2023.

Her optician in Bracknell, Berkshire, could not find anything wrong with her eyes, so, several weeks later, Jasmine was sent to hospital for an MRI brain scan.

Her father Anthony, 37, says he got the news that night: Jasmine had a brain tumour.

The next day, a neurosurgeon further explained to Anthony and Jakki, Jasmine's mother, that she had an incurable midline glioma.

'We were told that she likely only had 12 to 18 months even if she underwent radiotherapy,' says Anthony, a former operations manager at Heathrow airport, who is separated from Jakki. 'I refused to believe that there was simply nothing that they could do.'

Anthony researched experimental overseas treatments and found a drug called Onc-201 being tested in the US and the Netherlands. The family raised £246,000 via GoFundMe for the treatment but Jasmine died in January 2024, less than a year after diagnosis. Anthony says: 'We wanted to keep her with us as long as possible.'

https://www.dailymail.co.uk/health/article-13435169/truth-foreign-cancer-clinics-terminally-ill-child-experts-alarm-unproven-treatments.html

Pelizaeus-Merzbacher siblings

My name is Robert and I live in Franklin Square, New York with my family. My wife Kristen and I have four sons; Brian (27) Trevor (25), Andrew (22), and Dylan (20). All of them are currently living at home.

We had thought our life was changing when a sonogram in 1996 revealed that Brian would be born with a bi-lateral cleft lip and palate. Our research indicated that it could be fixed soon after birth and he would be able to live a normal life. Kristen and I were prepared to face that challenge. Brian was born on May 20 of that year at 41 weeks. We were not surprised when he was brought to the neonatal ICU shortly after birth.

Shortly on, we noticed that he had nystagmus, a shaking of the eyeballs. He never reached any developmental milestones such as XYZ. We had no idea what was wrong. This was the worst part. Neither of us has a family history of neurological disorders, although I had an aunt with a cleft palate. We went to several doctors in an attempt to find a diagnosis. We were first told that it could be a birth injury due to him being born one week late and having meconium in the amniotic fluid. That could have been aided by the bi-lateral cleft palate. We also tested for a multitude of other diseases. Brian was always happy and cognizant of everything around him. He just had poor head control and couldn’t drink or eat unassisted.

Brian received physical, occupational, and speech / feeding therapy. However, none of these therapists or the physicians knew what they were treating. They just considered it to be “cerebral palsy-like.)

This continued for the next seven years. We continued with our team of doctors for Brian. We were seeing a plastic surgeon and a prosthodontist for his face, and a neurologist, orthopedist, physiatrist, ophthalmologist, gastroenterologist, nutritionist, cardiologist, as well as regular visits to his pediatrician.

Kristen gave birth on June 26, 2003 to our 4th son Dylan. Its was her fourth Cesarean section. I was excited to meet the new member of the family. I just had a feeling in my gut that something wasn’t right by just looking at him. I’m not sure if it was parental instinct or the experience at Brian’s birth which I had suppressed, but I just noticed something was wrong.

I went over to Kristen and she asked about him while she was getting her tubes tied. I told her, “I think we might have another Brian on our hands.” Her look at me said it all

Dylan went into the neonatal ICU and was discharged with us a few days later. His neurological development mirrored Brian’s, but not their personalities. Brian was a lovable child that would smile at everyone that would come up to him. Dylan knew what he wanted and made sure you knew it, and had a sarcastic smile, even at a young age.




In 2004, My son’s neurologist, Dr. Chaula Kharode, took a blood sample that was sent to Baylor University for a F.I.S.H. (fluorescence in situ hybridization) test.

We got the positive results for PMD on a Saturday. Brian and I flew to Indianapolis the following Wednesday for a meeting of the PMD Family Support group. I was surprised to see adults with the disease, and meet some of the most knowledgeable, caring, and down to earth parents I have ever met. We felt at home and relieved that it was not a death sentence.

Fast forward to the present. The six of us are living with PMD. It affects the entire family. Brian and Dylan are reliant on us for all daily life skills. They cannot sit up or eat and drink unassisted. They cannot be left home alone. Trevor works in Connecticut but lives at home to help us out. It is increasingly difficult to lift them. Durable medical equipment has been able to help especially XYZ., . Brian was also hospitalized twice in 2023 for pulmonary issues, including getting a chest tube inserted.

Brian and Dylan both attended a special needs school with Brian graduating at age 21. He now attends a day program for disabled adults. They realize they are different from the abled population but that doesn’t affect their personalities. They enjoy travelling up to their grandparent’s lake house, going to hockey games, and meeting new people.

They each have their own choices in what television shows to watch, who sits where in the van, and which setting to have the air conditioner on. They are non-verbal, but we know what they want. They definitely let us know!

We started sending them to a summer sleep-away camp for the disabled in around 2016. They love it. Dylan will let out an angry cry when we come to visit, because he thinks we are picking him up. Camping trips have to be to accessible sites, it is difficult to go to events together as a family. We are happy to send them back to camp after a three-year hiatus. The staff at Camp Loyaltown, in Hunter, New York, is wonderful. We were only worried for them the first year.

Kristen and I are realists. Any new therapeutic that comes out will probably not affect our boys, but anything to keep them comfortable and extend their lives would have to be considered. I am more concerned about future generations. PMD in the Gasperetti family will end with this generation, as we do not have the girl we always wanted.

https://www.pmdfoundation.org/blog/gasperetti-brothers

Perplexity on my part regarding XYZ.

Next-generation phenotyping for patients with ultrarare disorders

Schmidt, A., Danyel, M., Grundmann, K. et al. Next-generation phenotyping integrated in a national framework for patients with ultrarare disorders improves genetic diagnostics and yields new molecular findings. Nat Genet (2024). https://doi.org/10.1038/s41588-024-01836-1

Abstract

Individuals with ultrarare disorders pose a structural challenge for healthcare systems since expert clinical knowledge is required to establish diagnoses. In TRANSLATE NAMSE, a 3-year prospective study, we evaluated a novel diagnostic concept based on multidisciplinary expertise in Germany. Here we present the systematic investigation of the phenotypic and molecular genetic data of 1,577 patients who had undergone exome sequencing and were partially analyzed with next-generation phenotyping approaches. Molecular genetic diagnoses were established in 32% of the patients totaling 370 distinct molecular genetic causes, most with prevalence below 1:50,000. During the diagnostic process, 34 novel and 23 candidate genotype–phenotype associations were identified, mainly in individuals with neurodevelopmental disorders. Sequencing data of the subcohort that consented to computer-assisted analysis of their facial images with GestaltMatcher could be prioritized more efficiently compared with approaches based solely on clinical features and molecular scores. Our study demonstrates the synergy of using next-generation sequencing and phenotyping for diagnosing ultrarare diseases in routine healthcare and discovering novel etiologies by multidisciplinary teams.
_______________________________________________________

The majority of rare diseases have a genetic cause. The underlying genetic alteration can be found more and more easily, for example, by means of exome sequencing (ES), leading to a molecular genetic diagnosis. ES is an examination of all sections of our genetic material (DNA) that code for proteins. As part of a Germany-wide multicenter study, ES data was collected from 1,577 patients and systematically evaluated.

This made it possible to diagnose a total of 499 patients, with 34 patients showing new, previously unknown genetic diseases. The study thus makes a significant contribution to the initial description of new diseases. In addition, software based on the use of artificial intelligence (AI) was used for the first time on a broad scale to support clinical diagnosis.

The "GestaltMatcher" AI system can assist in the assessment of facial features with regard to the classification of congenital genetic syndromes. The results of the study, in which 16 university locations were involved, have been published in Nature Genetics.

Ultra-rare diseases require both multidisciplinary clinical expertise and comprehensive genetic diagnostics for optimal care. The three-year TRANSLATE NAMSE innovation fund project began at the end of 2017 with the aim of improving the care of those affected by means of modern diagnostic concepts.

Researchers from 16 university hospitals analyzed the ES data of 1,577 patients, including 1,309 children, who presented to rare disease centers as part of TRANSLATE NAMSE. The aim of the project was to find the cause of the disease in as many patients as possible using innovative examination methods.

A genetic cause of the rare disease was identified in 499 patients, 425 of whom were children. In total, the researchers found changes in 370 different genes.

"We are particularly proud of the discovery of 34 new molecular diseases, which is a great example of knowledge-generating patient care at university hospitals," says Dr. Theresa Brunet, one of the lead authors from the Institute of Human Genetics at the Klinikum rechts der Isar of the Technical University of Munich.

What happens next with the unsolved cases?

"We will examine the affected patients for whom we have not yet been able to find a diagnosis as part of the model project Genome Sequencing, or MVGenomSeq for short," says Dr. Tobias Haack, Deputy Director of the Institute of Medical Genetics and Applied Genomics at the University Hospital of Tübingen.

The MVGenomSeq builds on the success of the TRANSLATE NAMSE project and enables the analysis of clinical genomes at university hospitals throughout Germany. Unsolved cases can also be investigated in follow-up studies using new examination methods, such as long-read sequencing, which allows much longer DNA fragments to be analyzed.

"Long-read sequencing enables us to find genetic changes that are difficult to detect and we assume that we will be able to make further diagnoses using this method," says Dr. Nadja Ehmke, Head of Genome Diagnostics at Charité's Institute of Medical Genetics and Human Genetics and one of the last authors.

As part of the TRANSLATE NAMSE project, standardized procedures for extended genetic diagnostics for suspected rare diseases were also established at the participating rare disease centers, based on interdisciplinary case conferences. These were incorporated into standard care after the project was completed.

"The interdisciplinary case conferences play an important role for those affected. This enables a comprehensive clinical characterization, which is relevant for the phenotype-based evaluation of the genetic data. In addition, the detected variants can be discussed in an interdisciplinary context," says Dr. Magdalena Danyel, one of the first authors, who works as a specialist at the Institute of Medical Genetics and Human Genetics and a fellow of the Clinician Scientist Program of the Berlin Institute of Health (BIH) at Charité—Universitätsmedizin.

Rare genetic diseases can sometimes be recognized by the face

The researchers also investigated whether the supplementary use of machine learning and artificial intelligence (AI) tools improves diagnostic effectiveness and efficiency.

To this end, the "GestaltMatcher" software developed by researchers in Bonn, which uses computer-assisted facial analysis to support the person using it in the diagnosis of rare diseases, was tested on a broad scale for the first time.

The study used the sequence and image data of 224 people who had also consented to the computer-assisted analysis of their facial images, and it was shown that the AI-supported technology provides a clinical benefit.

The GestaltMatcher AI can recognize abnormalities in the face and assign them to specific diseases. An important question when assessing genetic data is: Does the phenotype match the genotype? The AI can provide support here.

"GestaltMatcher is like an expert opinion that we can provide to any medical professional in a matter of seconds. Early diagnosis is essential for those affected by rare diseases and their families. Supportive use of the software by pediatricians could already be useful in the case of abnormalities during the U7 screening at 21 to 24 months or U7a at 34 to 36 months," says corresponding author Prof. Peter Krawitz, Director of the Institute for Genomic Statistics and Bioinformatics (IGSB) at the University Hospital Bonn (UKB), where the GestaltMatcher AI is being developed.

Prof. Krawitz is also a member of the Cluster of Excellence ImmunoSensation2 and in the Transdisciplinary Research Areas (TRA) "Modeling" and "Life & Health" at the University of Bonn. The software and app can be made available to all doctors through the non-profit organization Arbeitsgemeinschaft für Gen-Diagnostik e.V. (AGD).

https://medicalxpress.com/news/2024-07-genetic-diagnostics-ultra-rare-diseases.html




Wednesday, July 24, 2024

Congenital myasthenic syndromes 2

Nathalie Smeets, Alexander Gheldof, Bart Dequeker, , Margaux Poleur , Sofia Maldonado Slootjes, Vinciane Van Parijs, Nicolas Deconinck, Pauline Dontaine,  Alicia Alonso-Jimenez, Jan De Bleecker, Willem De Ridder, Sarah Herdewyn, Stéphanie Paquay, Arnaud Vanlander, Liesbeth De Waele, Diane Beysen, Kristl G. Claeys, Nicolas Dubuisson, Isabelle Hansen, Gauthier Remiche, Sara Seneca, Véronique Bissay, Luc Régal. Congenital Myasthenic Syndromes in Belgium: Genetic and Clinical Characterization of Pediatric and Adult Patients. Pediatric Neurology, 2024-09-01, Volume 158, Pages 57-65.

Abstract

Background

Congenital myasthenic syndromes (CMS) are a group of genetic disorders characterized by impaired neuromuscular transmission. CMS typically present at a young age with fatigable muscle weakness, often with an abnormal response after repetitive nerve stimulation (RNS). Pharmacologic treatment can improve symptoms, depending on the underlying defect. Prevalence is likely underestimated. This study reports on patients with CMS followed in Belgium in 2022.

Methods

Data were gathered retrospectively from the medical charts. Only likely pathogenic and pathogenic variants were included in the analysis.

Results

We identified 37 patients, resulting in an estimated prevalence of 3.19 per 1,000,000. The patients harbored pathogenic variants in CHRNE, RAPSN, DOK7, PREPL, CHRNB1, CHRNG, COLQ , MUSK, CHRND, GFPT1, and GMPPB. CHRNE was the most commonly affected gene. Most patients showed disease onset at birth, during infancy, or during childhood. Symptom onset was at adult age in seven patients, caused by variants in CHRNE , DOK7 , MUSK , CHRND, and GMPPB . Severity and distribution of weakness varied, as did the presence of respiratory involvement, feeding problems, and extraneuromuscular manifestations. RNS was performed in 23 patients of whom 18 demonstrated a pathologic decrement. Most treatment responses were predictable based on the genotype.

Conclusions

This is the first pooled characterization of patients with CMS in Belgium. We broaden the phenotypical spectrum of pathogenic variants in CHRNE with adult-onset CMS. Systematically documenting larger cohorts of patients with CMS can aid in better clinical characterization and earlier recognition of this rare disease. We emphasize the importance of establishing a molecular genetic diagnosis to tailor treatment choices.


Daniel Natera-de Benito, Alessia Pugliese, Kiran Polavarapu, Velina Guergueltcheva, Ivailo Tournev, Albena Todorova, Joana Afonso Ribeiro, Daniel M. Fernández-Mayoralas, Carlos Ortez, Loreto Martorell, Berta Estévez-Arias, Leslie Matalonga, Steven Laurie, Cristina Jou, Jarred Lau, Rachel Thompson, Xinming Shen, Andrew G. Engel, Andres Nascimento, Hanns Lochmüller,
Duygu Selcen. Advancing the Understanding of Vesicle-Associated Membrane Protein 1-Related Congenital Myasthenic Syndrome: Phenotypic Insights, Favorable Response to 3,4-Diaminopyridine, and Clinical Characterization of Five New Cases. Pediatric Neurology, 2024-08-01, Volume 157, Pages 5-13.

Abstract

Background

Congenital myasthenic syndromes (CMS) are a group of inherited neuromuscular junction (NMJ) disorders arising from gene variants encoding diverse NMJ proteins. Recently, the VAMP1 gene, responsible for encoding the vesicle-associated membrane protein 1 (VAMP1), has been associated with CMS.

Methods

This study presents a characterization of five new individuals with VAMP1-related CMS, providing insights into the phenotype.

Results

The individuals with VAMP1-related CMS exhibited early disease onset, presenting symptoms prenatally or during the neonatal period, alongside severe respiratory involvement and feeding difficulties. Generalized weakness at birth was a common feature, and none of the individuals achieved independent walking ability. Notably, all cases exhibited scoliosis. The clinical course remained stable, without typical exacerbations seen in other CMS types. The response to anticholinesterase inhibitors and salbutamol was only partial, but the addition of 3,4-diaminopyridine (3,4-DAP) led to significant and substantial improvements, suggesting therapeutic benefits of 3,4-DAP for managing VAMP1-related CMS symptoms. Noteworthy is the identification of the VAMP1 (NM_014231.5): c.340delA; p.Ile114SerfsTer72 as a founder variant in the Iberian Peninsula and Latin America.

Conclusions

This study contributes valuable insights into VAMP1-related CMS, emphasizing their early onset, arthrogryposis, facial and generalized weakness, respiratory involvement, and feeding difficulties. Furthermore, the potential efficacy of 3,4-DAP as a useful therapeutic option warrants further exploration. The findings have implications for clinical management and genetic counseling in affected individuals. Additional research is necessary to elucidate the long-term outcomes of VAMP1-related CMS.

Thursday, July 18, 2024

Novel whole genome sequencing approaches and lessons for neuropathies

Novel Whole Genome Sequencing Approaches and Lessons for Neuropathies

June 25, 2024

Technological advancements and cost efficiencies in whole genome sequencing are driving the discovery of repeat expansions associated with inherited neuropathies. Dr. Stephan Zuchner, Professor of Human Genetics and Neurology at the University of Miami Miller School of Medicine, highlighted the importance of long-range human genome sequencing at the 2024 Peripheral Nerve System (PNS) Annual Meeting in Montreal, Canada, noting that approaches like PacBio, Oxford Nanopore, and Optical Genome Mapping are crucial for evaluating neuropathy-associated structural variations. He pointed out the challenges of next-generation sequencing (NGS) technologies: a significant portion of variations fall outside the coding region, and half of our genome is comprised of repeat elements.

"By far, the largest group of variants are rare variants. So rare variants are actually the most common type of variant." Dr. Zuchner discussed significant advancements in genomic analysis for neurological disorders, emphasizing the potential of NGS technologies in identifying causative genes of peripheral neuropathies. He stressed the importance of understanding the genetic basis of neuropathies and the potential for clinical trials, noting that "what we see in inherited neuropathies might be quite meaningful for other types of peripheral neuropathies."

Dr. Zuchner also discussed the discovery of three causative genes of neuropathy where structural changes were identified through short-read sequencing. However, he explained, "If we had long-read sequencing, we would have discovered them much faster." These three genes were RFC1, SORD, and FGF14. RFC1 and FGF14 are linked to late-onset cerebellar ataxia, and SORD is involved in Charcot-Marie-Tooth disease and diabetic neuropathy. Further elaborating on genomic instability and its implications for therapy, Dr. Zuchner explained that the FGF14 GAA repeat expansion locus exhibits germline meiotic instability, with inheritance from parents leading to size changes. He discussed how a protective sequence in FGF14 prevents repeat instability. "We have discovered a 70-base pair sequence just in front of the FGF14 repeat that is completely protective of such dynamic expansion. We looked at thousands of long-range sequences, and we can see the complete discrimination between people who have this 70-base pair protective physical variant versus those who don't. It really determines if you will expect or you will not expect ataxia."

Dr. Zuchner noted the potential of using the latest advancements in artificial intelligence and machine learning to predict classified genome variants into binary pathogenic categories by treating DNA and resulting protein sequences like a language. Large language models can yield accurate classifications of protein variants and determine their pathogenicity by comparing sequence variations across different species.

This Omics session shed light on the critical role of long-range whole genome sequencing and artificial intelligence in evaluating structural variations and understanding the complexities of rare variants in neuropathies. It also highlighted the potential for developing targeted therapies based on genomic insights.

https://focusonneurology.com/pns2024/novel-whole-genome-sequencing-approaches-and-lessons-for-neuropathies




Congenital myasthenic syndromes

Kediha MI, Tazir M, Sternberg D, Eymard B, Ali Pacha L. Innovative Therapeutic Approaches in Congenital Myasthenic Syndromes. Neurol Clin Pract. 2024 Jun;14(3):e200277. doi: 10.1212/CPJ.0000000000200277. Epub 2024 May 7. PMID: 38737513; PMCID: PMC11081764.

Abstract

Background and objectives: To provide real-word clinical follow-up data on patients carrying variations of congenital myasthenic syndromes (CMS) and who respond to some innovative drugs.

Methods: Patients recruited from the Neurology Department of the Mustapha Bacha university hospital in Algiers. Treated with innovative drugs, they were monitored and their clinical progress was evaluated on the basis of clinical arguments suggestive of CMSs, but also para clinical arguments (electromyography and genetic study).

Results: Six patients carrying different mutations in different genes of CMSs were studied. They had different pathophysiologic profiles (slow or fast channel syndromes, low expressor of receptor). Their therapeutic management was based on innovative drugs, normally indicated in other, non-neurological pathologies. Their outcome was toward a clear clinical improvement.

Discussion: This work relates the interest of proposing treatments (outside of Pyridostigmine) in the management of CMSs. These therapies can greatly modify the prognosis of patients suffering from this orphan disease.

Classification of evidence: This study provides Class IV evidence that for patients with congenital myasthenic syndromes, some innovative treatments are effective.

Kao JC, Milone M, Selcen D, Shen XM, Engel AG, Liewluck T. Congenital myasthenic syndromes in adult neurology clinic: A long road to diagnosis and therapy. Neurology. 2018 Nov 6;91(19):e1770-e1777. doi: 10.1212/WNL.0000000000006478. Epub 2018 Oct 5. PMID: 30291185; PMCID: PMC6251603.

Abstract

Objective: To investigate the diagnostic challenges of congenital myasthenic syndromes (CMS) in adult neuromuscular practice.

Methods: We searched the Mayo Clinic database for patients with CMS diagnosed in adulthood in the neuromuscular clinic between 2000 and 2016. Clinical, laboratory, and electrodiagnostic data were reviewed.

Results: We identified 34 patients with CMS, 30 of whom had a molecular diagnosis (14 DOK7, 6 RAPSN, 2 LRP4, 2 COLQ, 2 slow-channel syndrome, 1 primary acetylcholine receptor deficiency, 1 AGRN, 1 GFPT1, and 1 SCN4A). Ophthalmoparesis was often mild and present in 13 patients. Predominant limb-girdle weakness occurred in 19 patients. Two patients had only ptosis. Age at onset ranged from birth to 39 years (median 5 years). The median time from onset to diagnosis was 26 years (range 4-56 years). Thirteen patients had affected family members. Fatigable weakness was present when examined. Creatine kinase was elevated in 4 of 23 patients (range 1.2-4.2 times the upper limit of normal). Repetitive nerve stimulation revealed a decrement in 30 patients. Thirty-two patients were previously misdiagnosed with seronegative myasthenia gravis (n = 16), muscle diseases (n = 15), weakness of undetermined cause (n = 8), and others (n = 4). Fifteen patients received immunotherapy or thymectomy without benefits. Fourteen of the 25 patients receiving pyridostigmine did not improve or worsen.

Conclusion: Misdiagnosis occurred in 94% of the adult patients with CMS and causes a median diagnostic delay of nearly 3 decades from symptom onset. Seronegative myasthenia gravis and muscle diseases were the 2 most common misdiagnoses, which led to treatment delay and unnecessary exposure to immunotherapy, thymectomy, or muscle biopsy.

Wednesday, July 17, 2024

Clobazam pathway, pharmacokinetics

Inspired by a patient with levels clobazam 397 desmethylclobazam < 200.

Courtesy of a colleague.

Huddart R, Leeder JS, Altman RB, Klein TE. PharmGKB summary: clobazam pathway, pharmacokinetics. Pharmacogenet Genomics. 2018 Apr;28(4):110-115. doi: 10.1097/FPC.0000000000000327. PMID: 29517622; PMCID: PMC5914180.

Background

Clobazam is an antiepileptic drug (AED) which has been approved by the FDA for use in the treatment of Lennox-Gastaut Syndrome in patients 2 years and older. Outside of the U.S., clobazam is also used as an adjunctive therapy for refractory epilepsy and to treat other epileptic syndromes and anxiety. It is a 1,5-benzodiazepine, distinguished by the presence of a nitrogen atom at positions 1 and 5 of the benzodiazepine ring rather than at positions 1 and 4 as seen in the 1,4-benzodiazepines (e.g. diazepam) . At the time of writing, clobazam is the only 1,5-benzodiazepine in clinical use. The main metabolite of clobazam, norclobazam, is also clinically active and exerts an antiepileptic effect.

Epileptic seizures are thought to be caused by excessive excitatory action potentials in neurons.  Clobazam and norclobazam exert their effects by binding to postsynaptic GABAA receptors in the brain, ultimately causing hyperpolarization of the neuron to create an inhibitory signal. This hyperpolarization increases the action potential threshold, thereby reducing the frequency of action potentials and the likelihood of seizures. This is discussed in greater detail in the Pharmacodynamics section below.

Clobazam is generally considered to cause fewer side-effects and cutaneous reactions, such as Stevens-Johnson Syndrome (SJS) or toxic epidermal necrolysis (TEN), at a lower rate than other AEDs, such as the 1,4-benzodiazepine carbamazepine. Cases of SJS/TEN have been reported in patients taking clobazam, often in combination with the 1,4-benzodiazepine lamotrigine and valproic acid. As lamotrigine is associated with a high risk of SJS/TEN and clobazam and lamotrigine are not thought to interact with each other, it seems likely that lamotrigine is inducing SJS/TEN in these cases.

The FDA has identified 21 cases of SJS/TEN from both the US and abroad, many of these in children. As a result, they have released a warning about the potential for clobazam to cause SJS/TEN at https://www.fda.gov/drugs/drugsafety/ucm377204.htm. However, it should be noted that, in 19 of these cases, the patient was taking one or more concomitant drugs which are also associated with SJS/TEN , many of which are themselves considered to be high risk for SJS/TEN. Indeed, a recent case-control study of 480 cases of SJS/TEN found no causal association between clobazam and SJS/TEN.

Monday, July 15, 2024

GATAD2B-associated neurodevelopmental disorder (GAND)

Inspired by a patient

Shieh C, Jones N, Vanle B, Au M, Huang AY, Silva APG, Lee H, Douine ED, Otero MG, Choi A, Grand K, Taff IP, Delgado MR, Hajianpour MJ, Seeley A, Rohena L, Vernon H, Gripp KW, Vergano SA, Mahida S, Naidu S, Sousa AB, Wain KE, Challman TD, Beek G, Basel D, Ranells J, Smith R, Yusupov R, Freckmann ML, Ohden L, Davis-Keppen L, Chitayat D, Dowling JJ, Finkel R, Dauber A, Spillmann R, Pena LDM; Undiagnosed Diseases Network; Metcalfe K, Splitt M, Lachlan K, McKee SA, Hurst J, Fitzpatrick DR, Morton JEV, Cox H, Venkateswaran S, Young JI, Marsh ED, Nelson SF, Martinez JA, Graham JM Jr, Kini U, Mackay JP, Pierson TM. GATAD2B-associated neurodevelopmental disorder (GAND): clinical and molecular insights into a NuRD-related disorder. Genet Med. 2020 May;22(5):878-888. doi: 10.1038/s41436-019-0747-z. Epub 2020 Jan 17. Erratum in: Genet Med. 2020 Apr;22(4):822. doi: 10.1038/s41436-020-0760-2. PMID: 31949314; PMCID: PMC7920571.

Abstract

Purpose: Determination of genotypic/phenotypic features of GATAD2B-associated neurodevelopmental disorder (GAND).

Methods: Fifty GAND subjects were evaluated to determine consistent genotypic/phenotypic features. Immunoprecipitation assays utilizing in vitro transcription-translation products were used to evaluate GATAD2B missense variants' ability to interact with binding partners within the nucleosome remodeling and deacetylase (NuRD) complex.

Results: Subjects had clinical findings that included macrocephaly, hypotonia, intellectual disability, neonatal feeding issues, polyhydramnios, apraxia of speech, epilepsy, and bicuspid aortic valves. Forty-one novelGATAD2B variants were identified with multiple variant types (nonsense, truncating frameshift, splice-site variants, deletions, and missense). Seven subjects were identified with missense variants that localized within two conserved region domains (CR1 or CR2) of the GATAD2B protein. Immunoprecipitation assays revealed several of these missense variants disrupted GATAD2B interactions with its NuRD complex binding partners.

Conclusions: A consistent GAND phenotype was caused by a range of genetic variants in GATAD2B that include loss-of-function and missense subtypes. Missense variants were present in conserved region domains that disrupted assembly of NuRD complex proteins. GAND's clinical phenotype had substantial clinical overlap with other disorders associated with the NuRD complex that involve CHD3 and CHD4, with clinical features of hypotonia, intellectual disability, cardiac defects, childhood apraxia of speech, and macrocephaly.

Vera G, Sorlin A, Delplancq G, Lecoquierre F, Brasseur-Daudruy M, Petit F, Smol T, Ziegler A, Bonneau D, Colin E, Mercier S, Cogné B, Bézieau S, Edery P, Lesca G, Chatron N, Sabatier I, Duban-Bedu B, Colson C, Piton A, Durand B, Capri Y, Perrin L, Wiesener A, Zweier C, Maroofian R, Carroll CJ, Galehdari H, Mazaheri N, Callewaert B, Giulianno F, Zaafrane-Khachnaoui K, Buchert-Lo R, Haack T, Magg J, Rieß A, Blandfort M, Waldmüller S, Horber V, Leonardi E, Polli R, Turolla L, Murgia A, Frebourg T, Lebre AS, Nicolas G, Saugier-Veber P, Guerrot AM. Clinical and molecular description of 19 patients with GATAD2B-Associated Neurodevelopmental Disorder (GAND). Eur J Med Genet. 2020 Oct;63(10):104004. doi: 10.1016/j.ejmg.2020.104004. Epub 2020 Jul 17. PMID: 32688057.

Abstract

De novo pathogenic variants in the GATAD2B gene have been associated with a syndromic neurodevelopmental disorder (GAND) characterized by severe intellectual disability (ID), impaired speech, childhood hypotonia, and dysmorphic features. Since its first description in 2013, nine patients have been reported in case reports and a series of 50 patients was recently published, which is consistent with the relative frequency of GATAD2B pathogenic variants in public databases. We report the detailed phenotype of 19 patients from various ethnic backgrounds with confirmed pathogenic GATAD2B variants including intragenic deletions. All individuals presented developmental delay with a median age of 2.5 years for independent walking and of 3 years for first spoken words. GATAD2B variant carriers showed very little subsequent speech progress, two patients over 30 years of age remaining non-verbal. ID was mostly moderate to severe, with one profound and one mild case, which shows a wider spectrum of disease severity than previously reported. We confirm macrocephaly as a major feature in GAND (53%). Most common dysmorphic features included broad forehead, deeply set eyes, hypertelorism, wide nasal base, and pointed chin. Conversely, prenatal abnormalities, non-cerebral malformations, epilepsy, and autistic behavior were uncommon. Other features included feeding difficulties, behavioral abnormalities, and unspecific abnormalities on brain MRI. Improving our knowledge of the clinical phenotype is essential for correct interpretation of the molecular results and accurate patient management.

Abad C, Robayo MC, Muñiz-Moreno MDM, Bernardi MT, Otero MG, Kosanovic C, Griswold AJ, Pierson TM, Walz K, Young JI. Gatad2b, associated with the neurodevelopmental syndrome GAND, plays a critical role in neurodevelopment and cortical patterning. Transl Psychiatry. 2024 Jan 18;14(1):33. doi: 10.1038/s41398-023-02678-x. PMID: 38238293; PMCID: PMC10796954.

Abstract

GATAD2B (GATA zinc finger domain containing 2B) variants are associated with the neurodevelopmental syndrome GAND, characterized by intellectual disability (ID), infantile hypotonia, apraxia of speech, epilepsy, macrocephaly and distinct facial features. GATAD2B encodes for a subunit of the Nucleosome Remodeling and Histone Deacetylase (NuRD) complex. NuRD controls transcriptional programs critical for proper neurodevelopment by coupling histone deacetylase with ATP-dependent chromatin remodeling activity. To study mechanisms of pathogenesis for GAND, we characterized a mouse model harboring an inactivating mutation in Gatad2b. Homozygous Gatad2b mutants die perinatally, while haploinsufficient Gatad2b mice exhibit behavioral abnormalities resembling the clinical features of GAND patients. We also observed abnormal cortical patterning, and cellular proportions and cell-specific alterations in the developmental transcriptome in these mice. scRNAseq of embryonic cortex indicated misexpression of genes key for corticogenesis and associated with neurodevelopmental syndromes such as Bcl11b, Nfia and H3f3b and Sox5. These data suggest a crucial role for Gatad2b in brain development.

Thursday, July 11, 2024

AI for PA

As health plans increasingly rely on technology to deny treatment, physicians are fighting back with chatbots that synthesize research and make the case.

At his rehabilitation medicine practice in Illinois, Dr. Azlan Tariq typically spent seven hours a week fighting with insurance companies reluctant to pay for his patients’ treatments.

He often lost.

There was the 45-year-old man who spent five months in a wheelchair while his insurer denied appeal after appeal for a prosthetic leg. Or the stroke survivor who was rehospitalized following a fall after his insurer determined his rehab “could be done at home.”

Over the course of Dr. Tariq’s 12-year career, these stories had become more common: The list of treatments that needed pre-approval from insurers seemed ever broadening, and the denials seemed ever rising.

So in an effort to spare his patients what he deemed subpar care, and himself mountains of paperwork, Dr. Tariq recently turned to an unlikely tool: generative A.I.

For a growing number of doctors, A.I. chatbots — which can draft letters to insurers in seconds — are opening up a new front in the battle to approve costly claims, accomplishing in minutes what years of advocacy and attempts at health care reform have not.

“We haven’t had legislative tools or policymaking tools or anything to fight back,” Dr. Tariq said. “This is finally a tool I can use to fight back.”

Doctors are turning to the technology even as some of the country’s largest insurance companies face class-action lawsuits alleging that they used their own technology to swiftly deny large batches of claims and cut off seriously ill patients from rehabilitation treatment.

Some experts fear that the prior-authorization process will soon devolve into an A.I. “arms race,” in which bots battle bots over insurance coverage. Among doctors, there are few things as universally hated.

“If you want to see a physician go apoplectic at a cocktail party, mention prior authorizations,” said Dr. Robert Wachter, the chair of the medicine department at the University of California, San Francisco.

The process was designed by insurance companies to keep health care costs down by reining in doctors’ use of unnecessary and expensive treatments.

But Dr. Jonathan Tward, a radiation oncologist based in Utah, said insurers often demanded so much exhaustive documentation and paperwork — even for standard cancer treatments — that he felt he was on the losing side of a “war of attrition.”

Doctors and their staff spend an average of 12 hours a week submitting prior-authorization requests, a process widely considered burdensome and detrimental to patient health among physicians surveyed by the American Medical Association.

With the help of ChatGPT, Dr. Tward now types in a couple of sentences, describing the purpose of the letter and the types of scientific studies he wants referenced, and a draft is produced in seconds.

Then, he can tell the chatbot to make it four times longer. “If you’re going to put all kinds of barriers up for my patients, then when I fire back, I’m going to make it very time consuming,” he said.

Dr. Tariq said Doximity GPT, a HIPAA-compliant version of the chatbot, had halved the time he spent on prior authorizations. Maybe more important, he said, the tool — which draws from his patient’s medical records and the insurer’s coverage requirements — has made his letters more successful.

Since using A.I. to draft prior-authorization requests, he said about 90 percent of his requests for coverage had been approved by insurers, compared with about 10 percent before.

Generative A.I. has been particularly useful for doctors at small practices, who might not ordinarily have time to appeal an insurer’s decision — even if they think their patient’s treatment will suffer because of it.

Nearly half of doctors surveyed by the A.M.A. said that when they didn’t appeal a claim denial, it was at least in part because they didn’t have the time or resources for the insurance company’s lengthy appeals process.

Dr. Michael Albert, an obesity medicine specialist in Oklahoma, said A.I. had enabled his small, resource-strapped telehealth practice to go from almost never appealing insurance denials to sending 10 to 20 appeals per week.

Now, Dr. Albert said he could “operate at the same level as companies that have essentially infinite resources.”

While A.I. is still primarily used by individual, tech-savvy doctors, a growing number of companies are trying to bring the technology into the mainstream.

Epic, one of the largest electronic health record companies in the country, has rolled out a prior-authorization tool that uses A.I. to a small group of physicians, said Derek De Young, a developer working on the product.

Several major health systems are piloting Doximity GPT, created to help with a number of administrative tasks including prior authorizations, a company spokeswoman said.

But the insurance companies aren’t sitting still, either. Chris Bond, a spokesman for America’s Health Insurance Plans, said insurers welcomed attempts to streamline the process, including those involving the “appropriate use of A.I.”

Dr. Jeff Levin-Scherz, a health policy expert at the Harvard T.H. Chan School of Public Health, said he believed that most health plans were at least evaluating how to use artificial intelligence in their claims review process, if they weren’t using it already.

At the same time, these A.I. tools may make it easier for ill-intentioned players to bill for medically unnecessary treatments, Dr. Levin-Scherz noted.

As doctors use A.I. to get faster at writing prior-authorization letters, Dr. Wachter said he had “tremendous confidence” that the insurance companies would use A.I. to get better at denying them.

“You have automatic conflict,” said Dr. Wachter, who wrote a book about digital technology in medicine. “Their A.I. will deny our A.I., and we’ll go back and forth.”

Dr. Wachter said he hoped that some day, with more advanced A.I. technology, insurers and providers could create a new system that relies less on sending letters back and forth and more on automation.

Insurance coverage could be automatically approved based on an algorithm’s analysis of the most up-to-date scientific literature, for example. An A.I. tool might also notify a doctor that the $100,000 drug she’s about to prescribe could be substituted with a similarly effective $250 pill.

Until then, many doctors are enjoying a newfound, if temporary, foothold in the battle over insurance claims.

“It is definitely an A.I. race,” Dr. Tariq said, “but I think it wouldn’t be fair if the physicians didn’t have A.I.”

https://www.nytimes.com/2024/07/10/health/doctors-insurers-artificial-intelligence.html?

Wednesday, July 10, 2024

Highly superior autobiographical memory (HSAM)

See: https://www.youtube.com/watch?v=5MlFheM5Tf8

https://www.youtube.com/watch?v=Qb0hP_Rrjbg

https://www.youtube.com/watch?v=n0RNVB7dMrw

https://www.youtube.com/watch?v=hpTCZ-hO6iI

Highly Superior Autobiographical Memory (HSAM) is a memory phenomenon first described by researchers at the Center for the Neurobiology of Learning and Memory at UC Irvine. Individuals with HSAM have a superior ability to recall specific details of autobiographical events, tend to spend a large amount of time thinking about their past and have a detailed understanding of the calendar and its patterns.

Initial Discovery: In 2006, Professor James McGaugh and colleagues reported the first known case of HSAM in a research participant known as “AJ”, later identified as Jill Price. When provided with a date, Jill could specify on which day of the week it fell and what she did that day. Since then, more individuals with this extraordinary ability have been identified (over 50 now), and CNLM researchers have been working to understand this mysterious ability. The condition was renamed to HSAM from its original label, hyperthymesia, which was used in the original report. Read all of the scientific reports here. The phenomenon and the work have been highlighted in numerous media venues.

Where we stand: So far, a number of other individuals have been identified with similar abilities and a number of other research groups have begun to explore this phenomenon. Memory for public events has typically been used as a screening criterion which is then followed by more rigorous memory testing. However, it is now clear that these tests rely heavily on expertise as well as the age of the participant. There is also a strong cultural bias in the questions typically asked.

Currently, we are developing new tools to probe this phenomenon that do not rely on individual expertise or explicit knowledge of public events. If you suspect you might have this ability, click here to take the screening.

So far, the studied conducted at UC Irvine suggest that individuals with HSAM have superior abilities in autobiographical memories but are no different from age- and sex-matched control participants on standard laboratory memory tests. Furthermore, MRI studies of their brain show preliminary evidence of specific regions and networks that may be different from control participants, although this is work that is still quite preliminary.

We have also identified several children with this ability, and are now conducting genetic studies in twins to understand the potential genetic basis of this ability. Genetic studies are conducted in collaboration with Dr. Rudy Tanzi's research group at Harvard. If you suspect that your child may have this ability, please fill out the screening.

SDAM: In 2015, the opposite memory phenotype was identified by colleagues at Baycrest's Rotman Research Institute in Toronto. Professor Brian Levine and colleagues described and named a condition known as Severely Deficient Autobiographical Memory (SDAM), which refers to a lifelong inability to vividly recollect or re-experience personal past events from a first-person perspective. We are now launching a collaboration with our colleagues at the Rotman to understand both extremes of autobiographical memory.

Research team at UCI: Several investigators at the CNLM, in collaboration with Dr. McGaugh, are working closely to understand the HSAM phenomenon and its underlying neural basis. Most notably, CNLM Director, Dr. Michael Yassa, and Dr. Craig Stark as well as members of their research teams.

Collaborators outside UCI: Collaborating teams with UCI investigators include Patrizia Campolongo at Sapienza Universita di Roma, Valerio Santangelo at University Degli Studi di Perugia, Roddy Roediger at Washington University in St. Louis, Aaron Mattfeld at Florida State University, and Rudi Tanzi and Jaehong Suh at Harvard University and Ruth Benca at UC Irvine.

Supporting the research: The study of HSAM could significantly inform on how the brain can optimize or enhance its processing of memories, knowledge which can be used to reverse engineer the process such that we can use it to prevent or treat memory disorders. However, this type of fundamental discovery process is rarely supported by federal funds. Click here to find out how you can support this research.

https://cnlm.uci.edu/hsam/

Monday, July 8, 2024

Neurological consequences of SARS-CoV-2 infections in the pediatric population

Casabianca M, Caula C, Titomanlio L and Lenglart L (2023) Neurological consequences of SARS-CoV-2 infections in the pediatric population. Front. Pediatr. 11:1123348. doi: 10.3389/fped.2023.1123348

COVID-19 in the pediatric population is mostly asymptomatic. However, 1 out of 5 children presents non-specific neurologic symptoms such as headache, weakness, or myalgia. Furthermore, rarer forms of neurological diseases are increasingly being described in association to a SARS-CoV-2 infection. Encephalitis, stroke, cranial nerves impairment, Guillain-Barré syndrome or acute transverse myelitis have been reported and account for around 1% of pediatric COVID-19 cases. Some of these pathologies may occur during or after the SARS-CoV-2 infection. The pathophysiological mechanisms range from direct invasion of the central nervous system (CNS) by SARS-CoV-2 itself to postinfectious immune-mediated CNS inflammation. In most cases, patients presenting neurological pathologies related to SARS-CoV-2 infection are at greater risk of life-threatening complications and should be closely monitored. Further studies are needed to acknowledge the potential long-term neurodevelopmental consequences of the infection.


Homicide trial

KINGSPORT – Six years ago, a Sullivan County Grand Jury met and indicted Cody R. Webb, of Kingsport, on charges of first-degree murder in the death of a 4-year-old girl.

Six years later, he’s still awaiting a trial.

“The biggest delay in this case was COVID,” said William Harper, deputy district attorney general. “It was set for last year, but the defense asked for records from several state’s witnesses that required multiple hearings over the last six months.”

Webb found himself indicted on the charge of first-degree murder in October 2018.

But the murder and his alleged involvement began three months earlier.

On Sunday morning, Aug. 5, 2018, Kingsport officers were called to a home in Colonial Heights, finding the deceased child.

The police department immediately launched a suspicious death investigation and sent the body to the ETSU Quillen College of Medicine for an autopsy.

After the autopsy, police classified it as a homicide.

Police never released the cause of death or Webb’s relationship to the child.

But answers may soon come. A jury trial has been set for November for Webb, who also faces charges of child neglect, child abuse, tampering with evidence and violation of probation.

Why has it taken so long to prosecute a man facing murder charges?

Harper said the culprit is the pandemic that shut courts down for months then barely heard cases for years.

The Tennessee Supreme Court issued its first order declaring a state of emergency for the judicial branch on March 13, 2020.

This order suspended all in-person judicial proceedings. While courts did not close, this greatly hampered the ability of the courts to handle cases, especially jury trials.

The Tennessee Supreme Court issued several other orders throughout 2020, culminating with one on Dec. 20, 2020 that extended a ban on in-person court proceedings through January 2021 and jury trials through February 2021.

On May 14, 2021, the Court loosened some of those COVID restrictions.

“Despite things opening back up there was a palpable hesitancy on conducting jury trials,” Harper said.

He said the backlog of cases that developed from 2020 to 2021 put a large strain on resources and squeezed time that might have otherwise been used on jury trials.

Many matters continued to be handled remotely.

The case should soon see an ending and Harper said the prosecution will be ready for trial in November.

“Delays are never good for the prosecution, but at this point I do not think that it will impact our case any,” he said.

https://www.timesnews.net/news/crime/six-years-later-man-still-awaits-trial-on-murder-charges/article_3ae4a600-ee97-11ee-8b45-93cb39b7702d.html




Child abuse testimony



In April 2017, six-week-old J.M. slept most of the day and vomited “a lot” that evening. Mother noticed that J.M.’s arms began shaking at various times. Assuming it was a stomach issue, Father went to the store to buy tea for J.M. Meanwhile, J.M.’s condition worsened. J.M. turned pale, started moaning, could not fully open her eyes, and her arms became stiff. After Father returned from the store, Mother and Father took J.M. to an urgent-care center where they waited more than 40 minutes for the doctor to evaluate her. Upon examination, the doctor told Mother and Father to immediately take J.M. to Phoenix Children’s Hospital (“PCH”). At PCH, a scan revealed that J.M had a large subdural hemorrhage on the left side of her brain and a smaller subdural hemorrhage on the right. She also had significant midline shift and herniation of her brain, meaning there was so much pressure in the brain that it started to shift out of its normal position. J.M. required emergency neurosurgery to relieve the pressure because it had become so great that her skull could no longer contain the brain and its contents without threatening her life. She also had diffused retinal hemorrhages (or bleeding) in all quadrants of the retina and all layers of the retina. Her head injuries negatively affected a multitude of systems in her body. Post-trauma, doctors diagnosed her with cerebral palsy because she had significant motor impairment. She also suffers from regular epileptic seizures and is blind. She now requires occupational therapy, feeding therapy, and 24-hour monitoring. After surgery, Dr. Melissa Jones, a pediatrician with a specialty in child abuse pediatrics, evaluated J.M. After reviewing the family’s medical history and J.M.’s birth records, Dr. Jones determined the injuries resulted from abusive head trauma and Mother and Father provided no alternative explanation for the cause of J.M.’s injuries...

Dr. Jones testified for DCS, opining that J.M.’s injuries resulted from nonaccidental trauma. She
added that J.M.’s lack of external injuries did not rule out abuse. Dr. Ruth Bristol, J.M.’s pediatric
neurosurgeon, testified that J.M.’s injuries were most likely caused by recent trauma. She also
testified that J.M. will likely require long-term, full-time care for the foreseeable future.

Mother and Father’s expert, Dr. Joseph Scheller, a pediatric neurologist with specialties
in pediatric neurology and neuroimaging, disagreed with the Department’s experts. He testified
that J.M.’s injuries resulted from a subdural hematoma at birth that began spontaneously rebleeding some weeks later, which in turn caused her retinal hemorrhages. He conceded that this occurrence would be “an unusual complication” and that no other non-traumatic medical condition could have caused J.M.’s injuries.

In turn, Drs. Jones and Bristol opined on Dr. Scheller’s conclusion, testifying that such 
an occurrence under the circumstances present with J.M. would be “very, very rare.” Dr. Jones
testified that “children [who] have spontaneous re-bleeding [also] have some other complicating
factor with their brain.” Dr. Bristol testified that in her experience as a pediatric neurosurgeon she
had “not seen a spontaneous re-bleed to that degree.” Dr. Jones opined that J.M.’s presentation
and injuries did not correspond to Dr. Scheller’s theory, particularly the diffuse nature of J.M.’s
retinal hemorrhages, which was consistent with “massive trauma with acceleration and
deceleration.” Regarding J.M.’s eye injuries, Dr. Jones stated that:

[T]here had to be [a] significant force that led to that pattern of retinal hemorrhages.
You can get retinal hemorrhages from many different causes, but the only times we see
[J.M.’s] pattern of retinal hemorrhages in the pediatric population is from abusive head
trauma, severe motor vehicle collisions or there’s some case reports of children who
have fallen out of two or three story windows onto concrete.

Dr. Jones specifically distinguished Dr. Scheller’s theory, testifying that “when the pressure is
high in the brain, you can get retinal hemorrhages,” but they are typically “in the . . . most 
recessed part of the retina . . . surrounding the optic nerve,” which was “not the same pattern
that [J.M.] had.”

https://www.azcourts.gov/LinkClick.aspx?fileticket=URGwG0LpDNc%3D&portalid=45

Gottesfeld update 5

Martin Gottesfeld, 39, is a human rights advocate and senior systems engineer born and raised in Andover, Massachusetts. He was arrested February 17, 2016, and prosecuted under the Computer Fraud and Abuse Act (CFAA) by controversial Boston U.S. Attorney Carmen Ortiz. Update–on June 9, 2023, Gottesfeld was transferred to home confinement.

Gottesfeld faced allegations that he conspired with members of the "hacktivist" group Anonymous in organizing an online sit-in of Boston Children's Hospital's public website. At the time, an international campaign was underway to stop the torture of 15-year-old Justina Pelletier and save her life.

Gottesfeld faced a maximum of 15 years in prison and $380,000 in restitution to Pelletier's tormentors. At trial, prosecutors failed to prove the moral core of their case against him, below.

Gottesfeld dedicated years of his life to fighting for the human rights of institutionalized children. Often he spent eight hours at his job, came home, and spent another eight hours campaigning against child abuse. He never profited from his activism. He and his wife opened their guest bedroom to survivors of the "troubled teen industry," powerful and politically-connected for-profit businesses with an extremely well-documented record of atrocities against children. That was before more than a dozen F.B.I. agents raided their home and took every computer he owned.

In 2018 holdover prosecutors from Ortiz's office failed to convince a jury that anything Gottesfeld did in defending Pelletier potentially affected the care of even a single patient anywhere. That is because, quite simply, his defense of Pelletier's life posed no such risks.

In contrast, it's easy to see how Pelletier suffered while Gottesfeld's prosecutors did absolutely nothing to protect her.

The trial judges assigned to Gottesfeld's case were deeply connected to Boston Children's Hospital and its parent Harvard Medical School.

The Honorable Magistrate Judge Marianne Bowler, for instance, signed and sealed the search warrant for Gottesfeld's home and denied him bail. She had started her career as a medical research assistant at Harvard Medical School. She was also married to Marc Pfeffer, a Harvard cardiology professor and cardiologist at Brigham and Women's Hospital. The Brigham and Women's Hospital cardiology department touts its close ties to Boston Children's on its website. Bowler was also director emerita of The Boston Foundation, which raised hundreds of thousands of dollars for Boston Children's Hospital, and also raised money for the Wayside Youth and Family Support Network, which detained Pelletier under the Boston Children's Hospital treatment plan when Pelletier's physical presence at the hospital proved too controversial for comfort.

Magistrate Bowler signed and sealed the warrant for Gottesfeld's home September 29, 2014. The first page of the application for that warrant, however, was dated the following day, September 30, 2014.

Gottesfeld's trial judge was The Honorable Nathaniel M. Gorton. At all relevant times Judge Gorton was a director of and shareholder in his family's for-profit business Slade Gorton & Co., Inc. Slade Gorton & Co., Inc. donated to Boston Children's Hospital, which thanked Slade Gorton & Co., Inc. for its donations on the very same website that Gottesfeld allegedly took down. During the Pelletier case, Boston Children's Hospital hid its donor lists to protect companies like Slade Gorton & Co., Inc. from economic and reputational harm from Boston Children's Hospital's handling of the case.

Also, both Judge Gorton and his brother Michael Gorton had served on boards at The New England Home for Little Wanderers. Boston Children's Hospital gave that home a $50,000 direct grant in the years before Gottesfeld's case. And B.C.H. and the home had partnered to divert juvenile psychiatric patients, like Pelletier, to outpatient settings.

During the trial but after jury selection, one of Gottesfeld's jurors self-identified as a former accountant for The New England Home for Little Wanderers. She expressly told Judge Gorton she was worried about "a mistrial" due to her background. Judge Gorton refused to excuse her from the case. Then, when the jury had deadlocked for days, Judge Gorton repeatedly told the jury it could not acquit Gottesfeld due to his "good motive." When prosecutors instructed the jury, "you must convict" Gottesfeld, Judge Gorton did not correct them.

Then, when a tearful juror came forward during deliberations, pleading not to convict Gottesfeld, and describing pressure from other jurors, Judge Gorton refused defense counsel's timely motion to inquire upon the jury as to whether undue pressure was being exerted during deliberations.

Neither Magistrate Bowler nor Judge Gorton recused themselves when defense counsel moved for new judges.

Once convicted solely of economic harm to organizations that left Pelletier wheelchair-bound for life, Judge Gorton sentenced Gottesfeld to 121 months in federal prison and over $400,000 in restitution to organizations financially connected to Judge Gorton himself or Magistrate Bowler.

Pelletier remains in a wheelchair, having been denied any official justice whatsoever for her trauma at Boston Children's Hospital.

https://freemartyg.com/martys-story

Marty Gottesfeld may be getting a new home.

For several years now, he has called the Communications Management Unit (CMU) inside the federal prison first in Terre Haute, Indiana and then in Marion, Illinois.

He may be on the move, but it’s not clear if he will stay inside a CMU.

The CMU was created by the Federal Bureau of Prisons (FBP) after 9/11.

Primarily, it houses Jihadis and other dangerous individuals; Marty is a hacktivist, a non-violent offender.

Gottesfeld was convicted in 2016 of redirecting all sorts of internet traffic so that it attacked the website of Boston Children’s Hospital.

He did this stunt, Marty stated, because Boston Children’s Hospital was involved at the time in the medical kidnapping of Justina Pelletier.

His motive made little difference, and he was given a ten-year sentence.

He’s scheduled to be released in 2024.

Since arriving at the CMU, he has caused the FBP all sorts of trouble by spilling the secrets from inside this unit.

CMU is a euphemism, of course, for a unit where communication is difficult.

Difficult but not impossible and Marty has been able to communicate and leak stories: like the murder of an insurance scammer by a Jihadi and a guard who engages in sexual harassment.

His time in the CMU may be coming to an end.

https://michaelvolpe.substack.com/p/marty-gottesfeld-bids-adieu

Thursday, July 4, 2024

Primary amebic meningoencephalitis 8

A 14-year-old boy in India died Thursday from an infection caused by a brain-eating amoeba he contracted while swimming in contaminated water.

The teen, named Midrul, had reportedly picked up the parasite while bathing in a pond in Kozhikode, Kerala, India Today reported. He was subsequently admitted to a local hospital on June 24 after contracting an infection.

Despite medics’ best efforts, he succumbed to his symptoms, marking the third amoeba-related fatality in the region in less than two months.

The other victims were a five-year-old girl from Malappuram on May 21, and a 13-year-old Kannur girl who passed on June 25, the Economic Times reported.

Midrul had reportedly been infected by Naegleria fowleri, the now-notorious brain-eating amoeba that swims up people’s noses and wreaks havoc upon their brains.

He had specifically suffered from primary amebic meningoencephalitis (PAM), a catastrophic condition that causes the destruction of brain tissue and the swelling of the brain, according to the Centers for Disease Control and Prevention.

This microscopic scourge is found worldwide in warm freshwater bodies, including lakes, rivers and even poorly-maintained swimming pools. It cannot survive in saltwater nor can it be spread from person to person.

Symptoms — which generally occur between one and 12 days after infection — initially comprise severe headache, fever, nausea and vomiting before progressing to a stiff neck, seizures, and coma.

The condition is fatal 97% of the time, with deaths usually occurring less than a week after the onset of initial symptoms.

To date, there have been only five recorded survivors of PAM in North America.

https://nypost.com/2024/07/04/lifestyle/brain-eating-amoeba-kills-teen-after-he-went-swimming-in-contaminated-water-third-death-in-2-months/

Star student Megan Ebenroth, 17, tragically died last month after contracting a rare, brain-eating amoeba while swimming in Georgia.

The death was reported last month, but the victim’s identity wasn’t confirmed until now.

“I’m still in shock,” the teen’s mother Chrissy Ebenroth told the Atlanta Journal-Constitution while discussing the “extraordinary” pupil’s passing. “But I can’t keep silent about her.”

Megan and several friends had reportedly gone swimming on July 11 in a lake near her home in Dearing, McDuffie County.

The high schooler had wanted to enjoy the last few weeks of summer before starting her senior year.

Four days later, Megan awoke with a severe headache, prompting her mother to drive her to the hospital.

Doctors diagnosed the youth with sinusitis, prescribed her an antibiotic and sent her home.

Unfortunately, the teen’s symptoms continued to worsen over the week as she began to experience fever, migraines and a loss of balance, WLTX reported.

“It was all such a blur because her mental state changed so drastically,” said Chrissy.

Left with little other recourse, the distraught parent drove her daughter to the hospital, where she was immediately intubated and placed into a medically-induced coma.

At one point, doctors opened up the patient’s skull to relieve the swelling on her brain.

“The stage we were at was not one Megan could come back from,” lamented her crestfallen mother, who still had no idea what was causing her daughter’s symptoms.

It was not until that Friday, July 21 that doctors suggested that her complications were caused by an infection of naegleria fowleri, the now-notorious brain-eating amoeba that swims up people’s noses and colonizes their brains.

Naegleria fowleri is insidious as it’s often not detected until it’s too late.

Specifically, it infects victims with primary amebic meningoencephalitis, a catastrophic condition that causes the destruction of brain tissue and the swelling of the brain, according to the Centers for Disease Control and Prevention.

Although found worldwide, the microscopic monster predominantly resides in warm freshwater, such as lakes and rivers and hot springs, and can even inhabit poorly-maintained swimming pools.

Naegleria fowleri cannot survive in salt water and cannot spread from one person to another.

Unfortunately, the prognosis is not good for victims of this amoebic infection, which is reportedly fatal 97% of the time.

As was the case with Megan, symptoms — which can begin between 1 and 12 days following infection — initially entail severe headache, fever, nausea and vomiting before progressing to a stiff neck, seizures, and coma.

Death usually occurs within five days.

The parasite resides in freshwater bodies around the world.

A day after getting diagnosed, Megan tragically passed away — 11 days after initially contracting the parasite.

Paul Johnson, the coroner for McDuffie County, later confirmed that the HS senior had died from the rare brain infection, marking Georgia’s sixth amoeba-related fatality since 1962.

State health officials neglected to disclose the location of the lake for fear of insinuating that that’s the only body of water where the parasite could be residing.

“It just doesn’t feel real,” Chrissy Ebenroth said while discussing her daughter’s passing. “It seems like she’s going to walk into my house at any moment. It just doesn’t feel like this has happened to us.”

Ebenroth had gone swimming in a lake with friends to enjoy her last few weeks of summer before starting her senior year.

This marked a tragic end for Megan, who is remembered by friends and family as a highly-motivated student with big dreams and a vibrant personality.

Ever since sixth grade, Megan dreamed of going to the University of Georgia, and she worked hard to achieve her goal by making straight As and becoming president of the Beta Club and vice president of the Spanish Club.

The studious scholar had recently joined her high school tennis team to help round out her resume.

Megan’s mom Chrissy described herself and her daughter as “best friends.”

She explained, “She would tell people I was her best friend, and I would say ‘Honey, I can’t be your best friend’ and about three weeks ago, she said ‘come on, Mom, you know I’m your best friend, and I said, ‘Yes baby, you are.'”

The teen’s funeral was held on July 26 at the family’s church, Fort Creek Baptist Church, in Dearing, Georgia.

In light of the tragedy, Chrissy hopes to spread awareness of the disease, which is often not diagnosed until it’s too late.

“Going forward, I want that to be one of my main focuses,” the determined guardian declared, “to find a way to diagnose this earlier.”

Part of the problem is that only a few labs in the US offer the specific tests capable of detecting the microbe.

Not to mention that it’s often mistaken for viral meningitis, a more common disease that has similar symptoms but is far more curable.

Of the 157 known amoeba infectees in the US from 1962-2022, only four people have survived.

Unfortunately, N. Fowleri cases could see a major uptick in the future because the microbe could be spreading due to climate change.

Scientists claim that warming temperatures are creating ideal conditions for the amoeba to increasingly thrive in bodies of water across the northern US.

https://nypost.com/2023/08/04/star-student-dies-from-brain-eating-amoeba/

Friday, June 28, 2024

CSNK2B-related neurodevelopmental syndrome (Poirier-Bienvenu) 2

Li D, Zhou B, Tian X, Chen X, Wang Y, Hao S, Zhang C, Hui L. Genetic analysis and literature review of a Poirier-Bienvenu neurodevelopmental syndrome family line caused by a de novo frameshift variant in CSNK2B. Mol Genet Genomic Med. 2024 Jan;12(1):e2327. doi: 10.1002/mgg3.2327. Epub 2023 Nov 30. PMID: 38037515; PMCID: PMC10767686.

Abstract

Background: Poirier-Bienvenu neurodevelopmental syndrome (POBINDS) is a rare autosomal dominant neurologic disorder caused by a heterozygous variant of CSNK2B, which is characterized by early onset epilepsy, hypotonia, varying degrees of intellectual disability (ID), developmental delay (DD), and facial dysmorphism. This study clarifies the molecular diagnosis and causative factors of a Chinese boy with POBINDS.

Methods: The clinical phenotypes and ancillary laboratory tests were collected and analyzed by trio whole exome sequencing (WES) and copy number variant sequencing (CNV-seq) in the follow-up proband's families. The candidate variant was validated by Sanger sequencing and bioinformatics software was used to further explore the effect of the de novo frameshift variant on the protein structure.

Results: The proband carries a de novo frameshift variant c.453_c.454insAC (p.H152fs*76) in CSNK2B. According to the ACMG genetic variant classification criteria and guidelines, the locus is a pathogenic variant (PVS1+PS2+PM2) and the associated disease was POBINDS. Protein structure prediction suggests significant differences in amino acid sequences before and after mutation.

Conclusion: A rare case of POBINDS caused by a novel frameshift variant in CSNK2B was diagnosed. The novel variant extends the variation spectrum of CSNK2B, which provides guidance for early clinical diagnosis, genetic counseling and treatment of this family. A review of the currently reported cases of POBINDS further enriches and summarizes the relationship between genotype and phenotype of POBINDS.

Trivisano M, Dominicis A, Stregapede F, Quintavalle C, Micalizzi A, Cappelletti S, Dentici ML, Sinibaldi L, Calabrese C, Terracciano A, Vigevano F, Novelli A, Specchio N. Refining of the electroclinical phenotype in familial and sporadic cases of CSNK2B-related Neurodevelopmental Syndrome. Epilepsy Behav. 2023 Oct;147:109436. doi: 10.1016/j.yebeh.2023.109436. Epub 2023 Sep 15. PMID: 37717460.

Abstract

CSNK2B encodes a regulatory subunit of casein kinase II, which is highly expressed in the brain. Heterozygous pathogenic variants in CSNK2B are associated with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS) (OMIM #618732), characterized by facial dysmorphisms, seizures, intellectual disability, and behavioral disturbances. We report ten new patients with CSNK2B-related Neurodevelopmental Syndrome associated with heterozygous variants of CSNK2B. In three patients, a pathogenic variant was inherited from an affected parent. We describe both molecular and clinical features, focusing on epileptic and neurodevelopmental phenotypes. The median age at follow-up was 8.5 years (range 21 months-42 years). All patients had epilepsy, with onset at a median age of 10.5 months range 6 days-10 years). Seizures were both focal and generalized and were resistant to anti-seizure medications in two out of ten patients. Six patients had mild to moderate cognitive delays, whereas four patients had no cognitive disability. Although all previously reported patients had a de novo CSNK2B pathogenic variant, here we report, for the first time, two familial cases of CSNK2B-related Neurodevelopmental Syndrome. We confirmed the highly variable expressivity of the disease among both interfamilial and intrafamilial cases. Furthermore, this study provides information about the long-term outcome in adult patients and underlines the importance of detailed family history collection before performing genetic testing in patients with epilepsy and neurodevelopmental disorders.


Thursday, June 27, 2024

Drive through awake kidney transplant

Kidney transplants are not uncommon — the kidney is actually the most frequently transplanted organ in the U.S. — but a Chicago man recently received his in a highly unusual way.

John Nicholas, 28, was awake during the entire procedure, which was performed on May 24 at Northwestern Medicine Hospital. He was discharged the very next day.

Doctors administered a spinal anesthesia shot — similar to what is used for a Cesarean section — instead of general anesthesia, according to a press release from the hospital.

"This is the first case at Northwestern Medicine where a patient was awake during an entire kidney transplant procedure and went home the next day, basically making this an outpatient procedure," said Satish Nadig, M.D., PhD, transplant surgeon and director of the Northwestern Medicine Comprehensive Transplant Center, in the release.

"Inside the operating room, it was an incredible experience being able to show a patient what their new kidney looked like before placing it inside the body," he added.

"It was incredibly simple and uneventful."

This type of "awake transplant" could reduce surgical risks and shorten the length of the patient’s hospitalization, doctors say.

"It was an incredible experience, being able to show a patient what their new kidney looked like before placing it inside the body."

It could also increase access to care for patients who are considered high-risk or have phobias surrounding general anesthesia.

He was considered an ideal patient for awake surgery due to his young age, his otherwise good health and his "eagerness to participate," per the release.

"It was a pretty cool experience to know what was happening in real time and be aware of the magnitude of what they were doing," said Nicholas in a statement to Northwestern.

"At one point during surgery, I recall asking, ‘Should I be expecting the spinal anesthesia to kick in?’ They had already been doing a lot of work and I had been completely oblivious to that fact. Truly, no sensation whatsoever."

Although Nicholas did receive sedation for comfort, he said he was still aware of what was going on.

"Especially when they called out my name and told me about certain milestones they had reached," he noted.

Nicholas walked out of the hospital on May 25, the day after surgery.

Nicholas’ kidney issues began at age 16 when he was diagnosed with Crohn’s disease, the release stated.

After a few years of managing the condition with medication, his kidney function worsened and he required a transplant.

Nicholas’ mother originally planned to donate her kidney, but a breast cancer diagnosis prevented her from doing so, the hospital relayed.

It was Nicholas’ best friend from elementary school, 29-year-old Pat Wise in Alexandria, Virginia, who ultimately donated the life-saving kidney.

Dr. Marc Siegel, clinical professor of medicine at NYU Langone Medical Center and a Fox News medical contributor, was not involved in the surgery but shared his input.

"My opinion is that in most cases, spinal anesthesia presents a reasonable alternative for those at high risk of general anesthesia complications," Siegel told Fox News Digital.

"Having said that, general anesthesia would be preferred whenever possible for major surgery like this," said Dr. Siegel, "because control of the airway and breathing is essential, and is much easier if the patient is asleep and on the ventilator."

The doctors at Northwestern credit the patient for helping to advance the field of transplant medicine.

"When John agreed to be the first known patient at Northwestern Medicine to undergo an awake kidney transplant and be discharged home the next day, he knew the benefits outweighed the risks, and … he’s now helping to move the field of transplantation forward," said Vinayak Rohan, M.D., transplant surgeon at Northwestern Memorial Hospital, in the release.

"He is an extremely compliant patient who was in tune with his body and willing to push the envelope," stressing that the patient had faith in the doctors and they, in turn, had faith in him.

The hospital now plans to establish the AWAKE Program (Accelerated Surgery Without General Anesthesia in Kidney Transplantation) for patients who want or need to pursue surgery without general anesthesia.

"It really opens up a whole new door and is another tool in our toolbelt for the field of transplantation," Nadig added.

https://www.foxnews.com/health/chicago-man-receives-kidney-transplant-fully-awake-goes-home-next-day

Posterior reversible encephalopathy syndrome in children with malignancies

Kiermasz A, Zapała M, Zwiernik B, Stręk-Cholewińska A, Machnikowska-Sokołowska M and Mizia-Malarz A (2023) Posterior reversible encephalopathy syndrome in children with malignancies – a single-center retrospective study. Front. Neurol. 14:1261075. doi: 10.3389/fneur.2023.1261075

Background: Posterior reversible encephalopathy syndrome (PRES) diagnosis relies on clinical and radiological characteristics. Clinical manifestations include focal neurologic deficits, hemiparesis, seizures with symptoms of intracranial hypertension, headache, nausea, vomiting, and visual field disturbances. The majority of patients have typical changes in magnetic resonance imaging. The epidemiology and outcomes of PRES in the pediatric cancer population have not been well described. Most of the available data are from retrospective analyses.

Objective: The aim of our study was to evaluate the clinical and radiological presentation as well as the outcome of PRES in children treated for cancers in a single center.

Methods: We analyzed data from 1,053 patients diagnosed with malignancies in a single center over 15 years to determine the incidence of PRES.

Results: 19/1053 (1.8%) patients developed PRES. The diagnosis was accompanied by a range of clinical symptoms including hypertension, seizures, altered mental status, and headaches. Magnetic resonance imaging was performed in all patients, and 14/19 (73.7%) exhibited typical findings consistent with PRES. Four patients (21.0%) required treatment in the Intensive Care Unit.

Conclusion:

1. Posterior reversible encephalopathy syndrome (PRES) is a rare but significant complication in children with cancer.

2. There is a clear need to establish clinical criteria for PRES to improve the diagnosis and treatment of patients with PRES, particularly in the pediatric oncological population.

3. Further studies are needed to identify the risk factors for recurrent PRES, particularly in pediatric cancer patients undergoing chemotherapy or immunosuppressive treatment.

Wednesday, June 26, 2024

Martin Pistorius 3

https://www.youtube.com/watch?v=JtLw5KXm09U

https://www.youtube.com/watch?v=WD1IX1AFRZg

https://www.youtube.com/watch?v=GaAp7Q-fFiw

See: https://childnervoussystem.blogspot.com/2018/07/martin-pistorius-2.html

https://childnervoussystem.blogspot.com/2015/04/an-aromatherapist-saw-something-in-his.html

Recommendations for the assessment and management of idiopathic intracranial hypertension in children

Amin S, Monaghan M, Forrest K, Harijan P, Mehta V, Moran M, Mukhtyar B, Muthusamy B, Parker A, Prabhakar P, Whitehouse WP, Krishnakumar D. Consensus recommendations for the assessment and management of idiopathic intracranial hypertension in children and young people. Arch Dis Child. 2024 May 9:archdischild-2023-326545. doi: 10.1136/archdischild-2023-326545. Epub ahead of print. PMID: 38724065.

Abstract

Background: Idiopathic intracranial hypertension (IIH) is a potentially disabling condition. There is a lack of evidence and national guidance on how to diagnose and treat paediatric IIH, leading to variation in clinical practice. We conducted a national Delphi consensus via the Children's Headache Network to propose a best-practice diagnostic and therapeutic pathway.

Methods: The Delphi process was selected as the most appropriate methodology for examining current opinion among experts in the UK. 104 questions were considered by 66 healthcare professionals, addressing important aspects of IIH care: assessment, diagnosis, treatment, follow-up and surveillance. General paediatricians, paediatric neurologists, ophthalmologists, opticians, neuroradiologists and neurosurgeons with a clinical interest or experience in IIH, were invited to take part.

Results: The Delphi process consisted of three rounds comprising 104 questions (round 1, 67; round 2, 24; round 3 (ophthalmological), 13) and was completed between March 2019 and August 2021. There were 54 and 65 responders in the first and second rounds, respectively. The Delphi was endorsed by the Royal College of Ophthalmologists, which engaged 59 ophthalmologists for round 3.

Conclusions: This UK-based Delphi consensus process reached agreement for the management of paediatric IIH and has been endorsed by the Children's Headache Network and more broadly, the British Paediatric Neurology Association. It provides a basis for a pragmatic clinical approach. The recommendations will help to improve clinical care while minimising under and over diagnosis.

From the article:

Management: therapeutic LP

CSF pressure, if over 28 cm CSF (21 mm Hg) should be reduced down to 20–25 cm CSF (15–18 mm Hg).

The maximum number of (therapeutic) LPs a patient should have over the course of their illness is five, as other therapeutic options should be instigated before this point.

Management: weight management

A dietetic service should be available for weight management for patients with IIH.

Overweight or obese patients with IIH should be referred to a dietician for weight management or to a weight management team.

Management: first-line therapy in patients with IIH who do not have visual impairment

There was no consensus on medical management but the most selected answer by 46% of responders was that acetazolamide should be considered for all patients as first-line medical treatment, regardless of their BMI. This did not reach the threshold for consensus. The leading answer after ‘acetazolamide’ (46%) or ‘other’ (26%) was ‘information, general advice, safety netting but no drug treatment for now’ (18%). A minority of respondents supported topiramate (4%), no intervention (4%) or surgical approaches (2%).

Management: first-line therapy in patients with new visual impairment/loss of vision

If a patient is taking acetazolamide, blood urea, electrolytes and bicarbonate levels should be checked. Bicarbonate should be corrected when the value is equal to or lower than 18 mmol/L.

Management: second line

Repeated therapeutic LP should be offered when visual changes progress and there is a threat to vision on medical management and/or when symptoms are not responsive to medical management.

Management: neurosurgical

ICP bolt monitoring for 48 hours should be considered for patients with persistently raised LP opening pressure measurements (on two LPs) and papilloedema.

VSS should be considered if there is evidence of stenosis of the dominant venous sinus.




Recommendations for the assessment and management of Individuals With CDKL5 deficiency disorder

Amin S, Monaghan M, Aledo-Serrano A, Bahi-Buisson N, Chin RF, Clarke AJ, Cross JH, Demarest S, Devinsky O, Downs J, Pestana Knight EM, Olson H, Partridge CA, Stuart G, Trivisano M, Zuberi S, Benke TA. International Consensus Recommendations for the Assessment and Management of Individuals With CDKL5 Deficiency Disorder. Front Neurol. 2022 Jun 20;13:874695. doi: 10.3389/fneur.2022.874695. PMID: 35795799; PMCID: PMC9251467.

Abstract

CDKL5 Deficiency Disorder (CDD) is a rare, X-linked dominant condition that causes a developmental and epileptic encephalopathy (DEE). The incidence is between ~ 1:40,000 and 1:60,000 live births. Pathogenic variants in CDKL5 lead to seizures from infancy and severe neurodevelopmental delay. During infancy and childhood, individuals with CDD suffer impairments affecting cognitive, motor, visual, sleep, gastrointestinal and other functions. Here we present the recommendations of international healthcare professionals, experienced in CDD management, to address the multisystem and holistic needs of these individuals. Using a Delphi method, an anonymous survey was administered electronically to an international and multidisciplinary panel of expert clinicians and researchers. To provide summary recommendations, consensus was set, a priori, as >70% agreement for responses. In the absence of large, population-based studies to provide definitive evidence for treatment, we propose recommendations for clinical management, influenced by this proposed threshold for consensus. We believe these recommendations will help standardize, guide and improve the medical care received by individuals with CDD.

From the article:

There were many areas of consensus recommendations identified. The majority of these are for completion at baseline. There is an emphasis upon holistic care, such as the monitoring of systemic functions and educational needs, with certain areas recommended to be reviewed, not only at baseline, but also annually and if clinically indicated. These included the monitoring of growth, the need for a regular review of feeding and swallowing, and non-specialist screening for respiratory difficulties.

A comprehensive neurological assessment is encouraged at baseline. The consensus recommendations are for the individual with CDD to be reviewed by a pediatric neurologist with experience in managing epilepsy, clinician discussion to inform families about the risk of SUDEP, completion of a baseline MRI and EEG, consideration for epilepsy surgery, screening for the presence of a movement disorder, registration with the CDKL5 international registry and a review of the individual's sleep. Despite limited published evidence on the use of novel antiseizure drugs for CDD in the literature, Ganaxolone and Epidiolex are encouraged to be offered for epilepsy associated with CDD, if clinically indicated, dependent on FDA and EMA approvals and legal and regulatory requirements, respectively.

Discussion

CDD is a debilitating condition where there is an urgent need for further development of management options. To achieve these necessary advances will require large scale and international, collaborative efforts to evaluate potentially effective interventions in sufficiently powered clinical trials. Progress will rely heavily on cooperation between international medical and scientific professionals, affected families, industry and funding organizations. The extensive experience of the author group includes those with direct experience in CDD management including authors of a clinically relevant CDD severity assessment tool. We hope that this survey adds to the current knowledge base concerning clinical aspects of care and provides a useful proposed standard of care elucidated by the agreed areas of consensus. These recommendations can support clinicians with less experience of CDD and act as a catalyst for further research that would aim to increase capacity for evidence-based management in CDD.

Dell'Isola GB, Antonella F, Francesco P, Mario M, Cordelli DM, Piero P, Pasquale P, Alessandro F, Operto FF, Maurizio E, Marco C, Dario P, Sara M, Elisabetta S, Alberto S, Giovanna S, Savasta S, Paolo P, Di Cara G, Fruttini D, Vincenzo S, Pasquale S, Alberto V. CDKL5 deficiency-related neurodevelopmental disorders: a multi-center cohort study in Italy. J Neurol. 2024 Jun 14. doi: 10.1007/s00415-024-12421-1. Epub ahead of print. PMID: 38874638.

Abstract

CDKL5 deficiency disorder (CDD) is a complex clinical condition resulting from non-functional or absent CDKL5 protein, a serine-threonine kinase pivotal for neural maturation and synaptogenesis. The disorder manifests primarily as developmental epileptic encephalopathy, with associated neurological phenotypes, such as hypotonia, movement disorders, visual impairment, and gastrointestinal issues. Its prevalence is estimated at 1 in 40,000-60,000 live births, and it is more prevalent in females due to the lethality of germline mutations in males during fetal development. This Italian multi-center observational study focused on 34 patients with CDKL5-related epileptic encephalopathy, aiming to enhance the understanding of the clinical and molecular aspects of CDD. The study, conducted across 14 pediatric neurology tertiary care centers in Italy, covered various aspects, including phenotypic presentations, seizure types, EEG patterns, treatments, neuroimaging findings, severity of psychomotor delay, and variant-phenotype correlations. The results highlighted the heterogeneity of seizure patterns, with hypermotor-tonic-spasms sequence seizures (HTSS) noted in 17.6% of patients. The study revealed a lack of clear genotype-phenotype correlation within the cohort. The presence of HTSS or HTSS-like at onset resulted a negative prognostic factor for the presence of daily seizures at long-term follow-up in CDD patients. Despite extensive polypharmacotherapy, including medications such as valproic acid, clobazam, cannabidiol, and others, sustained seizure freedom proved elusive, affirming the inherent drug-resistant nature of CDD. The findings underscored the need for further research to explore response rates to different treatments and the potential role of non-pharmacological interventions in managing this challenging disorder.

Benke TA, Demarest S, Angione K, Downs J, Leonard H, Saldaris J, Marsh ED, Olson H, Haviland I. CDKL5 Deficiency Disorder. 2024 Apr 11. In: Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993–2024. PMID: 38603524.

Excerpt

Clinical characteristics: CDKL5 deficiency disorder (CDD) is a developmental and epileptic encephalopathy (DEE) characterized by severe early-onset intractable epilepsy and motor, cognitive, visual, and autonomic disturbances. Movement disorders include chorea, dystonia, and stereotypical hand and leg movements.

Although females are more commonly affected than males (female-to-male ratio is approximately 4:1), the severity of manifestations in heterozygous females and hemizygous males can be equivalent. However, the severity of the phenotype can vary depending on the type and position of the CDKL5 pathogenic variant, pattern of X-chromosome inactivation in females, and presence of postzygotic mosaicism in males or females, who can have mild manifestations.

Diagnosis/testing: The diagnosis of CDD is established in a female proband with suggestive clinical findings and a heterozygous CDKL5 pathogenic variant identified by molecular genetic testing.

The diagnosis of CDD is established in a male proband with suggestive clinical findings and a hemizygous CDKL5 pathogenic variant identified by molecular genetic testing.

Management: Treatment of manifestations: International consensus recommendations for the assessment and management of individuals with CDD have been published. The management of individuals with CDD is complex and requires multiple specialty evaluations; referral to a CDKL5 Center of Excellence may allow families to coordinate care more easily for affected individuals.

Targeted therapy: Ztalmy® (ganaxolone) is a targeted therapy for the treatment of epilepsy associated with CDD in individuals aged two years and older. This is the first approved treatment for seizures associated with CDD and the first treatment specifically for CDD.

Supportive care: Multidisciplinary care by specialists in the fields of pediatric neurology including pediatric epilepsy, feeding and nutrition, sleep disorders, behavioral disorders, orthopedics, physical therapy, occupational therapy, speech-language disorders, and genetic counseling.

Surveillance: Annual assessments by a medical home / primary care physician and specialists.

Genetic counseling: CDD is inherited in an X-linked manner. Approximately 99% of affected individuals represent simplex cases (i.e., a single occurrence in the family). The majority of individuals who represent simplex cases have the disorder as the result of a de novo germline or (rarely) postzygotic CDKL5 pathogenic variant. Rarely, an individual with CDD has the disorder as the result of a CDKL5 pathogenic variant inherited from a heterozygous or mosaic mother. If the mother of the proband has a CDKL5 pathogenic variant, the chance of transmitting it in each pregnancy is 50%. Females who inherit the pathogenic variant will be heterozygous and are at high risk of being affected, although skewed X-chromosome inactivation and the possibility of other attenuating factors may result in a variable phenotype. Males who inherit the pathogenic variant will be hemizygous and will most likely be severely affected. Once the CDKL5 pathogenic variant has been identified in an affected family member, prenatal and preimplantation genetic testing are possible.

See: https://childnervoussystem.blogspot.com/2019/10/cdkl5-genetic-disorder.html