2022

Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS. New England Journal of Medicine, 2022

VALOR and OLE Working Group, Sep 22 2022, In: New England Journal of Medicine. 387, 12, p. 1099-1110 12 p.

Antisense Oligonucleotides for the Study and Treatment of ALS. Neurotherapeutics, 2022

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by motor neuron loss. ALS is now associated with mutations in numerous genes, many of which cause disease in part through toxic gain-of-function mechanisms. Antisense oligonucleotides (ASOs) are small sequences of DNA that can reduce expression of a target gene at the post-transcriptional level, making them attractive for neutralizing mutant or toxic gene products. Advancements in the medicinal chemistries of ASOs have improved their pharmacodynamic profile to allow safe and effective delivery to the central nervous system. ASO therapies for ALS have rapidly developed over the last two decades, and ASOs that target SOD1, C9orf72, FUS, and ATXN2 are now in clinical trials for familial or sporadic forms of ALS. This review discusses the current state of ASO therapies for ALS, outlining their successes from preclinical development to early clinical trials.

Boros BD, Schoch KM, Kreple CJ, Miller TM. Antisense Oligonucleotides for the Study and Treatment of ALS. Neurotherapeutics. 2022 Jun 2. doi: 10.1007/s13311-022-01247-2. Epub ahead of print. PMID: 35653060.

Astrocytic 4R tau expression drives astrocyte reactivity and dysfunction. JCI Insight, 2022

To examine an astrocyte-specific role for 4R tau in disease, we used tau isoform-switching ASOs, previously characterized in our lab, to alter the ratio of tau isoform expression in hTau-expressing mouse and human astrocytes. Our findings demonstrate that, despite the low level of tau expression in astrocytes, 4R tau interferes with normal astrocyte functions and induces neuronal hyperexcitability.

Ezerskiy LA, Schoch KM, Sato C, Beltcheva M, Horie K, Rigo F, Martynowicz R, Karch CM, Bateman RJ, Miller TM. Astrocytic 4R tau expression drives astrocyte reactivity and dysfunction. JCI Insight. 2022 Jan 11;7(1):e152012. doi: 10.1172/jci.insight.152012. PMID: 34874917; PMCID: PMC8765054

2020

CNS microRNA profiles: a database for cell type enriched microRNA expression across the mouse central nervous system. Scientific Reports, 2020

In this manuscript, we detail the creation and possible uses of a new website that illustrates microRNA expression by cell type in the central nervous system. Similar databases for protein-coding genes have achieved widespread use, however no comparable tool for microRNA expression existed before this. We believe this new resource will serve as a valuable tool for researchers moving forward in microRNA research and enable the generation of previously unexplored hypotheses. This work was a collaboration between our group and the lab of Dr. Joseph Dougherty in the Departments of Genetics and Psychiatry at Washington University.

Pomper N, Liu Y, Hoye ML, Dougherty JD, Miller TM. CNS microRNA profiles: a database for cell type enriched microRNA expression across the mouse central nervous system. Sci Rep. 2020 Mar 18;10(1):4921. doi: 10.1038/s41598-020-61307-5. PMID: 32188880; PMCID: PMC7080788.

A Viral Toolkit for Recording Transcription Factor-DNA Interactions in Live Mouse Tissues. Proceedings of the National Academy of Sciences, 2020

In this manuscript, we describe the development of a new tool called “AAV calling cards” for measuring enhancer and transcription factor (TF)-mediated regulation in the mouse brain. It offers novel advantages over other methodologies, including the ability to probe epigenetic profiles in specific, targeted cellular populations and to record and integrate transient TF binding events over time. AAV calling cards will now enable unique studies in the areas of neurodevelopment and neurodegeneration. This work was a collaboration between our group and two labs in the Department of Genetics at Washington University, those of Dr. Robi Mitra and Dr. Joseph Dougherty.

Cammack AJ, Moudgil A, Chen J, Vasek MJ, Shabsovich M, McCullough K, Yen A, Lagunas T, Maloney SE, He J, Chen X, Hooda M, Wilkinson MN, Miller TM, Mitra RD, Dougherty JD. A viral toolkit for recording transcription factor-DNA interactions in live mouse tissues. Proc Natl Acad Sci U S A. 2020 May 5;117(18):10003-10014. doi: 10.1073/pnas.1918241117. Epub 2020 Apr 16. PMID: 32300008; PMCID: PMC7211997.

2019

Prospective natural history study of C9orf72 ALS clinical characteristics and biomarkers. Neurology Journal, 2019

2018

Protein production is an early biomarker for RNA-targeted therapies, 2018

Self WK, Schoch KM, Alex J, Barthélemy N, Bollinger JG, Sato C, Cole T, Kordasiewicz HB, Swayze E, Bateman RJ, Miller TM. Protein production is an early biomarker for RNA-targeted therapies. Ann Clin Transl Neurol. 2018 Oct 14;5(12):1492-1504. doi: 10.1002/acn3.657. PMID: 30564616; PMCID: PMC6292185.

Antisense oligonucleotides extend survival and reverse decrement in muscle response in ALS models. The Journal of Clinical Investigation, 2018.

Mutations in superoxide dismutase 1 (SOD1) are responsible for 20% of familial ALS. Given the gain of toxic function in this dominantly inherited disease, lowering SOD1 mRNA and protein is predicted to provide therapeutic benefit. We have developed next-generation SOD1 ASOs that more potently reduce SOD1 mRNA and protein and extend survival in SOD1G93A mice. These results define a highly potent, new SOD1 ASO ready for human clinical trial and suggest that at least some components of muscle response can be reversed by therapy.

McCampbell A, Cole T, Wegener AJ, Tomassy GS, Setnicka A, Farley BJ, Schoch KM, Hoye ML, Shabsovich M, Sun L, Luo Y, Zhang M, Comfort N, Wang B, Amacker J, Thankamony S, Salzman DW, Cudkowicz M, Graham DL, Bennett CF, Kordasiewicz HB, Swayze EE, Miller TM. Antisense oligonucleotides extend survival and reverse decrement in muscle response in ALS models. J Clin Invest. 2018 Aug 1;128(8):3558-3567. doi: 10.1172/JCI99081. Epub 2018 Jul 16. PMID: 30010620; PMCID: PMC6063493.

Motor neuron-derived microRNAs cause astrocyte dysfunction in amyotrophic lateral sclerosis. Brain, 2018.

We recently demonstrated that microRNA-218 (miR-218) is greatly enriched in motor neurons and is released extracellularly in amyotrophic lateral sclerosis model rats. To determine if the released, motor neuron-derived miR-218 may have a functional role in amyotrophic lateral sclerosis, we examined the effect of miR-218 on neighbouring astrocytes. Surprisingly, we found that extracellular, motor neuron-derived miR-218 can be taken up by astrocytes and is sufficient to downregulate an important glutamate transporter in astrocytes [excitatory amino acid transporter 2 (EAAT2)]. The effect of miR-218 on astrocytes extends beyond EAAT2 since miR-218 binding sites are enriched in mRNAs translationally downregulated in amyotrophic lateral sclerosis astrocytes. Inhibiting miR-218 with antisense oligonucleotides in amyotrophic lateral sclerosis model mice mitigates the loss of EAAT2 and other miR-218-mediated changes, providing an important in vivo demonstration of the relevance of microRNA-mediated communication between neurons and astrocytes. These data define a novel mechanism in neurodegeneration whereby microRNAs derived from dying neurons can directly modify the glial phenotype and cause astrocyte dysfunction.

Hoye ML, Regan MR, Jensen LA, Lake AM, Reddy LV, Vidensky S, Richard JP, Maragakis NJ, Rothstein JD, Dougherty JD, Miller TM. Motor neuron-derived microRNAs cause astrocyte dysfunction in amyotrophic lateral sclerosis. Brain. 2018 Sep 1;141(9):2561-2575. doi: 10.1093/brain/awy182. PMID: 30007309; PMCID: PMC6113638.

2017

Antisense oligonucleotides: Translation from mouse models to human neurodegenerative diseases. Neuron, 2017.

Multiple neurodegenerative diseases are characterized by single-protein dysfunction and aggregation.  Antisense oligonucleotides (ASOs) are small sequences of DNA able to target RNA transcripts, resulting in reduced or modified protein expression. ASOs hold considerable promise for neurodegenerative disease therapies as evidenced by SMA and ALS human clinical trial successes, and preclinical efforts in other CNS disorders continue to progress.

Schoch KM, Miller TM. Antisense Oligonucleotides: Translation from Mouse Models to Human Neurodegenerative Diseases. Neuron. 2017 Jun 21;94(6):1056-1070. doi: 10.1016/j.neuron.2017.04.010. PMID: 28641106; PMCID: PMC5821515.

MicroRNA Profiling Reveals Marker of Motor Neuron Disease in ALS Models. The Journal of Neuroscience, 2017.

Motor neurons are the cell type that is selectively lost in ALS. To develop tools to understand and monitor ALS disease progression, Mariah Hoye of Dr. Miller’s group identified factors that were specifically expressed in motor neurons only. These factors, called microRNAs (or miRNAs), may help researchers understand why motor neurons, and not other cell types, are selectively lost in ALS. One of the motor neuron enriched miRNAs was detected in spinal tap biofluid from an ALS rat model and its levels changed as disease progressed. These data suggest that miR-218 could be a clinically useful marker of disease status. Furthermore, Hoye found that rats treated with an ALS therapy had restored levels of the motor neuron marker, suggesting that miR-218 is a motor neuron specific and drug responsive marker for ALS rodents.

Hoye ML, Koval ED, Wegener AJ, Hyman TS, Yang C, O’Brien DR, Miller RL, Cole T, Schoch KM, Shen T, Kunikata T, Richard JP, Gutmann DH, Maragakis NJ, Kordasiewicz HB, Dougherty JD, Miller TM. MicroRNA Profiling Reveals Marker of Motor Neuron Disease in ALS Models. J Neurosci. 2017 May 31;37(22):5574-5586. doi: 10.1523/JNEUROSCI.3582-16.2017. Epub 2017 Apr 17. PMID: 28416596; PMCID: PMC5452343.

Tau Reduction Prevents Neuronal Loss and Reverses Pathological Tau Deposition and Seeding in Mice with Tauopathy. Science Translational Medicine, 2017.

Aggregates of a protein called “tau” are one of the factors involved in causing Alzheimer’s disease and other dementias. In this manuscript, Dr. Miller and colleagues show that lowering levels of tau protein prevented loss of neurons and reversed previous build up of tau protein in mouse models of Alzheimer’s disease. Additionally, using the same tau lowering strategy, researchers saw similar results when the drug was given to non-human primates. This study was the first to show that markers of neurodegeneration that accumulate in Alzheimer’s disease may ractually be reversed by treatment with antisense oligonucleotides.

DeVos SL, Miller RL, Schoch KM, Holmes BB, Kebodeaux CS, Wegener AJ, Chen G, Shen T, Tran H, Nichols B, Zanardi TA, Kordasiewicz HB, Swayze EE, Bennett CF, Diamond MI, Miller TM. Tau reduction prevents neuronal loss and reverses pathological tau deposition and seeding in mice with tauopathy. Sci Transl Med. 2017 Jan 25;9(374):eaag0481. doi: 10.1126/scitranslmed.aag0481. PMID: 28123067; PMCID: PMC5792300.

2016

Defining SOD1 ALS Natural History to Guide Therapeutic Clinical Trial Design. Journal of Neurology, Neurosurgery, and Psychiatry, 2016.

Natural history data is often used to help researchers understand disease characteristics and can be especially beneficial when considering how to design therapeutic clinical trials for a disease. In this manuscript, Dr. Miller’s group examined the natural history of ALS patients with mutations in the SOD1 gene in an attempt to provide updated information from a previous study that dated back to 1997. 15 medical centers contributed data from more than 175 ALS-SOD1 patients, and various disease measures such as age of onset were examined. The researchers concluded that the updated data was primarily unchanged from the previous study, showing support for the use of ALS natural history data in the design and implementation of clinical trials in the ALS-SOD1 patient population.

Bali T, Self W, Liu J, Siddique T, Wang LH, Bird TD, Ratti E, Atassi N, Boylan KB, Glass JD, Maragakis NJ, Caress JB, McCluskey LF, Appel SH, Wymer JP, Gibson S, Zinman L, Mozaffar T, Callaghan B, McVey AL, Jockel-Balsarotti J, Allred P, Fisher ER, Lopate G, Pestronk A, Cudkowicz ME, Miller TM. Defining SOD1 ALS natural history to guide therapeutic clinical trial design. J Neurol Neurosurg Psychiatry. 2017 Feb;88(2):99-105. doi: 10.1136/jnnp-2016-313521. Epub 2016 Jun 3. PMID: 27261500; PMCID: PMC5136332.

Increased 4R-Tau Induces Pathological Changes in a Human-Tau Mouse Model. Neuron, 2016.

Aggregates of a protein called “tau” are one of the factors involved in causing Alzheimer’s disease and other dementias. However, the tau protein has several different forms and it was not known which form of the protein was more detrimental and thus which form could be targeted for therapy in Alzheimer’s disease. In this manuscript, Dr. Kathleen Schoch of Dr. Miller’s group demonstrated, for the first time, that the “4R” form of tau was more toxic in mouse models of Alzheimer’s disease. Additionally, she showed that 4R tau could be effectively reduced in mice, indicating that a similar approach to lowering toxic 4R tau could be used to treat Alzheimer’s disease and other tauopathies in future studies.

Schoch KM, DeVos SL, Miller RL, Chun SJ, Norrbom M, Wozniak DF, Dawson HN, Bennett CF, Rigo F, Miller TM. Increased 4R-Tau Induces Pathological Changes in a Human-Tau Mouse Model. Neuron. 2016 Jun 1;90(5):941-7. doi: 10.1016/j.neuron.2016.04.042. Epub 2016 May 19. PMID: 27210553; PMCID: PMC5040069.

2015

In vivo kinetic approach reveals slow SOD1 turnover in the CNS. The Journal of Clinical Investigation, 2015.

Mutations in the SOD1 gene are known to cause some forms of familial ALS. Researchers are developing a treatment to reduce the level of SOD1 in familial ALS, but it is essential to understand how long SOD1 stays in the body to determine if the new treatment is effective in reducing SOD1. This manuscript aimed to determine the half-life of the protein in the cerebral spinal fluid using a new mass spectrometry technique called silk-isotope labeling kinetics (SILK). Researchers found that in rat models of ALS, SOD1 is a long-lived protein with a similar half-life in the cerebral spinal fluid and in the central nervous system. When applied to human participants, the SILK method worked successfully and confirmed the long half-life of SOD1. This study provided important insights into the kinetics of SOD1 and will help to develop future interventions for ALS.

Crisp MJ, Mawuenyega KG, Patterson BW, Reddy NC, Chott R, Self WK, Weihl CC, Jockel-Balsarotti J, Varadhachary AS, Bucelli RC, Yarasheski KE, Bateman RJ, Miller TM. In vivo kinetic approach reveals slow SOD1 turnover in the CNS. J Clin Invest. 2015 Jul 1;125(7):2772-80. doi: 10.1172/JCI80705. Epub 2015 Jun 15. PMID: 26075819; PMCID: PMC4563686.

2013

Method for widespread microRNA-155 inhibition prolongs survival in ALS-model mice. Human Molecular Genetics, 2013.

MicroRNAs are small molecules located throughout the body that can regulate various processes inside cells. For this reason, they are being investigated as potential therapeutic targets in a variety of diseases. In this manuscript, Dr. Miller’s group identified microRNAs that were changed in a rat model of ALS compared to normal rats. Six of these microRNAs were also found to be changed in human ALS tissues. The researchers then developed antisense oligonucleotide (ASO) inhibitors of the microRNA in order to test whether decreasing levels of one of the microRNAs, miR-155, would be beneficial in the ALS model. It was discovered that the ASO drug inhibited miR-155 across the brain and caused an increase in survival by 10 days and disease duration by 15 days (38%) versus control animals. This study provided support for the use of ASOs to successfully inhibit microRNAs throughout the brain and spinal cord as a way to treat ALS.

Koval ED, Shaner C, Zhang P, du Maine X, Fischer K, Tay J, Chau BN, Wu GF, Miller TM. Method for widespread microRNA-155 inhibition prolongs survival in ALS-model mice. Hum Mol Genet. 2013 Oct 15;22(20):4127-35. doi: 10.1093/hmg/ddt261. Epub 2013 Jun 4. PMID: 23740943; PMCID: PMC3781640.

Antisense reduction of tau in adult mice protects against seizures. Journal of Neuroscience, 2013.

Tau is a protein that forms aggregates in Alzheimer’s disease and causes hyperexcitability in neurons, which contributes to disease progression. Previous studies have shown that genetic deletion of tau substantially reduces hyperexicitability in various mouse models of Alzheimer’s disease, induced seizure models, and genetic models of epilepsy. However, researchers in Dr. Miller’s lab wanted to test a more translatable method of tau reduction in adult animals in order to determine whether this strategy could be applied to human patients. They found that antisense oligonucleotide (ASO)-mediated tau reduction decreased tau expression throughout the mouse central nervous system and caused less severe seizures compared to control mice. These results demonstrate that tau reducing ASOs could benefit those with epilepsy and potentially other disorders associated with tau-mediated neuronal hyperexcitability.

DeVos SL, Goncharoff DK, Chen G, Kebodeaux CS, Yamada K, Stewart FR, Schuler DR, Maloney SE, Wozniak DF, Rigo F, Bennett CF, Cirrito JR, Holtzman DM, Miller TM. Antisense reduction of tau in adult mice protects against seizures. J Neurosci. 2013 Jul 31;33(31):12887-97. doi: 10.1523/JNEUROSCI.2107-13.2013. PMID: 23904623; PMCID: PMC3728694.

An antisense oligonucleotide against SOD1 delivered intrathecally for patients with SOD1 familial amyotrophic lateral sclerosis: a phase 1, randomised, first-in-man study. Lancet Neurology, 2013.

Dr. Miller and colleagues present the results of the first-in-man Phase I clinical trial of antisense oligonucleotides (ASOs) targeted to SOD1 in familial ALS patients in this publication. Mutations in the gene SOD1 cause approximately 13% of genetically inherited ALS. ASOs targeting SOD1 prolonged survival in preclinical animal studies. The goal of this clinical trial was to assess the safety, tolerability, and pharmacokinetics of an SOD1 ASO (ISIS 333611) after intrathecal administration in patients with SOD1 related ALS. No dose-limiting toxic effects or any safety or tolerability concerns related to ISIS 333611 were noted and no serious adverse events occurred in patients given the drug. This was the first clinical study of intrathecal delivery of an ASO, and the researchers concluded that ISIS 333611 was well-tolerated when administered as an intrathecal infusion. Additional clinical trials are now underway.

Miller TM, Pestronk A, David W, Rothstein J, Simpson E, Appel SH, Andres PL, Mahoney K, Allred P, Alexander K, Ostrow LW, Schoenfeld D, Macklin EA, Norris DA, Manousakis G, Crisp M, Smith R, Bennett CF, Bishop KM, Cudkowicz ME. An antisense oligonucleotide against SOD1 delivered intrathecally for patients with SOD1 familial amyotrophic lateral sclerosis: a phase 1, randomised, first-in-man study. Lancet Neurol. 2013 May;12(5):435-42. doi: 10.1016/S1474-4422(13)70061-9. Epub 2013 Mar 29. Erratum in: Lancet Neurol. 2013 May;12(5):423. PMID: 23541756; PMCID: PMC3712285.