Industry partners from the companies noted below worked closely with the Healey & AMG Center Trial Design Team, the Northeast ALS Consortium (NEALS), Barrow Neurological Institute and Berry Consultants to tailor the arms of the HEALEY ALS Platform Trial to their experimental study drugs. Visit the trial page on ClinicalTrials.gov for more information about sites and trial participation criteria.

Download General Brochure

Biomarker Brochure

Now Enrolling

Regimen F: ABBV-CLS-7262, by Calico and AbbVie- Now Recruiting 

ABBV-CLS-7262 is an investigational drug developed by Calico Life Sciences LLC in collaboration with AbbVie Inc.  ABBV-CLS-7262 aims to restore function in cells affected by ALS by normalizing protein synthesis and preventing further sequestration and aggregation of TDP-43, thereby protecting neurons, and possibly slowing ALS progression.

The integrated stress response (ISR) is a fundamental transient process that regulates cell function during various stressful conditions.  Tissue studies suggest that the ISR is chronically induced in people with ALS.  It is proposed that TDP-43 aggregates, a hallmark feature in the motor neurons of people with ALS, could be formed by a chronically induced ISR.  ABBV-CLS-7262 activates the protein complex eIF2B, which is a key regulator of the ISR.  Binding of ABBV-CLS-7262 desensitizes eIF2B to stress and decreases the ISR.  Reduction of the ISR restores normal protein synthesis, reduces TDP-43 sequestration in stress granules, and may decrease TDP-43 aggregation.

A prior first-in-human study of ABBV-CLS-7262 showed that this drug was well-tolerated by participants, demonstrated target engagement by increasing eIF2B enzymatic activity, and suppressed the ISR in blood cells.  ABBV-CLS-7262 crossed the blood brain barrier at concentrations predicted to be efficacious in ALS.  ABBV-CLS-7262 is currently being investigated in a Phase 1b study in people with ALS (NCT04948645), and will be studied further as part of the HEALEY ALS Platform Trial.

Watch a webinar about the science behind ABBV-CLS-7262

Watch this video for more information on the mechanism of action behind ABBV-CLS-7262. 

Download Regimen F Brochure
Download Lumbar Puncture Brochure
Step by Step Guide to Lumbar Punctures: Watch short video<
Regimen G: DNL343, by Denali Therapeutics- Now Recruiting 

DNL343 is an investigational drug developed by Denali Therapeutics Inc. DNL343 aims to improve survival of nerve cells and slow ALS progression by restoring normal protein production and decreasing potentially harmful buildup of TDP-43 in cells affected by ALS. Abnormal TDP-43 buildup in nerve cells is found in >95% of individuals living with ALS.

The integrated stress response (ISR) appears to be overactive in ALS, and chronic activation of the ISR can lead to cellular dysfunction. In stressed cells, eIF2B activity is suppressed by the ISR, which leads to impaired protein synthesis and the formation of stress granules containing TDP-43. TDP-43 containing stress granules are thought to lead to TDP-43 inclusions, a hallmark of ALS pathology. DNL343 is a drug that is designed to inhibit the ISR, restore normal protein synthesis, and prevent the formation of TDP-43 containing stress granules as well as dissolve existing ones; the effects of which may be beneficial in the treatment of ALS.

Prior studies of DNL343 showed that this drug is generally well tolerated in healthy participants and individuals living with ALS. Experimental treatment with DNL343 showed a reduction in ISR biomarkers measured in the blood, suggesting that DNL343 inhibits the ISR. Analysis of cerebrospinal fluid (the fluid that surrounds the brain, spinal cord, and nerve cells impacted by ALS) obtained from participants in these studies showed that DNL343 is well distributed in the spinal fluid. Results from previous studies support continued evaluation of DNL343 as a potential treatment for ALS in the HEALEY ALS Platform Trial. DNL343 is also being studied in an ongoing Phase 1b trial (NCT05006352) in people with ALS.

Download Regimen G Brochure

Watch a webinar about the science behind DNL343

No Longer Enrolling

Regimen A: Zilucoplan, by UCB - Enrollment Complete

Zilucoplan is a small macrocyclic peptide inhibitor of complement component 5 [C5]. The complement system, as part of the innate immune system, is a group of proteins that evolved to protect humans from bacterial infections. In many diseases, inappropriate complement activation and deposition can occur damaging tissues and organs. Robust experimental data supports a role for complement activation and deposition in both the peripheral and central nervous systems in amyotrophic lateral sclerosis (ALS).

Zilucoplan, a new drug candidate developed by UCB (formerly Ra Pharmaceuticals), binds complement component 5 (C5) and inhibits tissue damage caused by pathological complement activation. Clinical data in a Phase 2 trial of zilucoplan in another neuromuscular condition generalized myasthenia gravis, demonstrated rapid, clinically meaningful and statistically significant improvements, as well as a favorable safety and tolerability profile. Zilucoplan has been designed for convenient “in-home” use by patients and is self-administered as a small volume, subcutaneous administration.

Watch a webinar about the science behind zilucoplan.

Note: On March 1, 2022, the zilucoplan regimen was halted.

Regimen B: Verdiperstat, by Biohaven Pharmaceutical Holding Company Ltd. – Enrollment Complete
Verdiperstat is an oral myeloperoxidase inhibitor. Microglia are the primary immune cells of the central nervous system. The presence of large numbers of activated microglia is one of the hallmarks of neurodegeneration in ALS. Myeloperoxidase (MPO) is a powerful pro-oxidant enzyme that is present in activated immune cells, including microglia. MPO is believed to increase oxidative stress and inflammation levels in the brain and spinal cord. Inhibiting MPO is anticipated to ameliorate these pathological mechanisms, which are strongly implicated in the onset and progression of ALS. Verdiperstat (BHV-3241) is a potential first-in-class, potent, selective, brain-permeable, irreversible myeloperoxidase enzyme inhibitor. Verdiperstat has been studied in approximately 250 people in Phase 1 and 2 clinical trials. It was generally safe and well-tolerated and has shown the potential to reduce microglial activation.
Watch a webinar about the science behind verdiperstat.

Note:
On Sept 29, 2022, the topline results for verdiperstat were announced
Regimen C: Bioenergetic Nanocatalysis (CNM-Au8, nanocrystalline gold) by Clene Nanomedicine, Inc. – Enrollment Complete

Motor neurons consume significant energy in order to function normally. In ALS, corrupted energy metabolism together with increased cellular stress lead to motor neuron degeneration. CNM-Au8 is a new class of medicine that provides an energetic assist to impaired motor neurons, helping them improve their ability to function more normally. CNM-Au8 acts catalytically to both support bioenergetic reactions inside cells and eliminate the harmful waste byproducts of cellular metabolism. Oral delivery of CNM-Au8 resulted in both neuroprotection and remyelination in multiple animal studies. Each 2 oz dose of CNM-Au8 is a concentrated, liquid suspension of pure gold nanocrystals that study participants drink every morning. These extremely small nanocrystals travel through the body and enter the brain and motor neuron cells where they enhance the ability of these cells to survive and communicate by supporting cellular metabolism. CNM-Au8 was demonstrated to be safe and well-tolerated by healthy volunteers in a Phase 1 study.

Watch a webinar about the science behind CNM-Au8.

Note:
On Oct 3, 2022, the topline results for CNM-Au8 were announced

Regimen D: Pridopidine, by Prilenia Therapeutics – Enrollment Complete

Pridopidine is a highly selective Sigma-1 receptor (S1R) agonist developed by Prilenia for the treatment of neurodegenerative and neurodevelopmental disorders. S1R regulates key cellular pathways, commonly impaired in neurodegeneration. Of particular interest is its role in the pathogenesis of ALS which is supported by human genetic and postmortem studies as well as by preclinical models. Pridopidine demonstrates robust neuroprotective effects in numerous preclinical models of neurodegenerative diseases including models of ALS. Compelling preclinical data supports the therapeutic potential of pridopidine in ALS. In ALS SOD1G93A motor neurons (MNs), pridopidine exerts neuroprotective effects via activation of the S1R. Specifically, pridopidine increases MN survival, improves BDNF and GDNF axonal transport, and restores the neuro-muscular junction (NMJ) synaptic activity. In vivo, pridopidine treatment of SOD1G93A mice reduces toxic protein aggregates and ameliorates muscle fiber wasting.

Clinical support for the validity of the S1R as a potential target for ALS can be derived from a prior trial using a non-selective S1R agonist, showing S1R activation may enhance bulbar and speech function in ALS patients. Pridopidine in vivo target engagement in humans is validated by PET imaging, providing support of the optimal clinical dose. Prior clinical data with pridopidine demonstrates a favorable safety and tolerability profile and provides evidence for a beneficial effect on functional outcome in another neurodegenerative disease.

Watch a webinar about the science behind pridopidine.

Previous research supports pridopidine’s neuroprotective properties in Huntington’s disease models. For more information, you can view information on pridopidine in Huntington's disease or read a selection of published articles about pridopidine.

Note:
On Feb 23, 2023, the topline results for pridopidine were announced

Download a brochure about Regimens A-D

Regimen E: Trehalose (SLS-005), by Seelos Therapeutics – Enrollment Complete

Trehalose is a low molecular weight disaccharide (0.342 kDa) that crosses the blood-brain barrier, stabilizes proteins and, importantly, activates autophagy, which is the process that clears pathologic material from cells. In animal models of ALS, treatment with trehalose delayed onset of disease, prolonged survival and preserved motor function and motor neurons in the spinal cord. Trehalose activates autophagy through the activation of Transcription Factor EB (TFEB), a key factor in lysosomal and autophagy gene expression. Activation of TFEB is an emerging therapeutic target for a myriad of diseases with pathologic accumulation of storage material. The drug is manufactured by Seelos Therapeutics, Inc., a clinical-stage biopharmaceutical company focused on the development of therapies for central nervous system disorders and rare diseases.

Webinar: The Science Behind Trehalose.

Webinar: Healey ALS Platform Trial Regimen E: A Participant’s Guide

Webinar: Update on Healey ALS Platform Trial Regimen E: Trehalose for ALS

Download brochure