logo
Latus Bio Unveils AAV-Ep+ Capsid Variant Capable of Unprecedented Protein Production in the Brain

Latus Bio Unveils AAV-Ep+ Capsid Variant Capable of Unprecedented Protein Production in the Brain

Business Wire14-05-2025
PHILADELPHIA--(BUSINESS WIRE)-- Latus Bio, Inc. (Latus), a biotechnology company pioneering advances in AAV gene therapy, has announced new research published today in Science Translational Medicine, 'AAVs engineered for robust brain transduction drive therapeutically relevant expression of secreted recombinant protein in NHPs and a mouse model of lysosomal storage disease.'
"This breakthrough in AAV capsid engineering represents a critical advancement in the field of gene therapy," said Dr. Beverly Davidson, Chair of the Scientific Advisory Board of Latus Bio and corresponding author of the study.
The study, led by Latus founder Beverly Davidson, PhD details the development of a novel adeno-associated virus (AAV) capsid variant - AAV-Ep+ - that demonstrates unprecedented potency in transducing cells that line the ventricles, known as ependymal cells, and cerebral neurons in mice and in non-human primates (NHPs). This advancement is potentially a significant leap forward for therapeutic gene delivery, wherein the study authors demonstrate that cells transduced with AAV-Ep+ can effectively serve as protein production depots, secreting large amounts of soluble proteins into the cerebral spinal fluid (CSF) for uptake throughout the central nervous system (CNS). This potency and distribution profile could potentially result in one-time delivery of gene therapies that encode protein treatments for lysosomal storage disorders (LSDs) as well as for other neurodevelopmental and neurodegenerative diseases that result in long term benefits for patients.
The AAV-Ep+ capsid variant was identified through a massively parallelled and unbiased screen of a large-diversity AAV variant library administered to NHPs. The nominated capsid, which was isolated from tens of millions of potential candidates, displays:
Remarkable tropism for cells that line the ventricular system of the brain and spinal cord of adult NHPs and mice. It also efficiently transduces neurons in cortical regions of the brain that are implicated in many diseases.
Robust transduction of induced pluripotent stem cell (iPSC)-derived human neurons and mice when compared to naturally occurring AAV serotypes. This cross-species activity highlights the potential for AAV-Ep+ to deliver sustained and therapeutic expression of encoded proteins in human brain cells that could result in prolonged therapeutic benefit for patients.
Low dose administration of AAV-Ep+ constructs designed to express human tripeptidyl peptidase (hTPP1) to mice deficient in TPP1 (a model of human CLN2 disease - a type of LSD) as well as to NHPs result in CSF and parenchymal tissue levels that exceeded those obtained with natural serotype capsids, reaching levels that are potentially multi-fold above therapeutic values required for CLN2 patients.
"This breakthrough in AAV capsid engineering represents a critical advancement in the field of gene therapy," said Dr. Beverly Davidson, Chair of the Scientific Advisory Board of Latus Bio and corresponding author of the study. "AAV-Ep+ offers a highly efficient, low-dose solution for brain-wide protein delivery, opening new possibilities for treating neurodevelopmental diseases like CLN2 disease and beyond."
The study showcases Latus' unique capsid discovery platform and ability to identify AAV capsid variants that are optimized for delivery to specific tissues and cell types, seeking to address translational shortcomings to prospectively enable better gene therapies. Latus continues to advance its pipeline of novel AAV capsid variants that target disease-relevant cell types in other regions of the central nervous system (e.g., cortex, cerebellum, spinal cord) as well as in peripheral tissues (e.g., ear, eye, heart, kidney and muscle). The Company is developing cutting-edge gene therapies that aim to transform the treatment landscape of genetically defined diseases, including many with high unmet medical needs.
About Latus Bio (Latus)
Latus is a biotechnology company dedicated to addressing devastating CNS and peripheral diseases via gene therapy. The Company is advancing an innovative therapeutics pipeline based on novel AAV capsid variants with potency and specificity. Latus is powered by a diverse team of visionary scientists, experienced clinicians, and leading industry executives. The Company has offices in Philadelphia, PA and in the Seaport in Boston, MA.
For more information, visit www.latusbio.com and follow on LinkedIn.
Orange background

Try Our AI Features

Explore what Daily8 AI can do for you:

Comments

No comments yet...

Related Articles

Could a single shot at birth shield kids from HIV for years?
Could a single shot at birth shield kids from HIV for years?

UPI

time5 days ago

  • UPI

Could a single shot at birth shield kids from HIV for years?

There's potentially exciting news from a trial conducted in monkeys: A single shot of gene therapy given to newborn monkeys appears to shield them from HIV, the virus that causes AIDS, for at least three years. Of course, studies conducted in animals don't always pan out in humans. But scientists say that if it does, it could save the lives of babies and children still imperiled by HIV. The study authors estimate that more than 100,000 children worldwide (largely in subSaharan Africa) are believed to contract HIV soon after birth, primarily via breastfeeding with an HIV+ mother. "Nearly 300 children are infected with HIV each day," said lead author Dr. Amir Ardeshir, associate professor of microbiology and immunology at the Tulane National Primate Research Center in New Orleans. "This approach could help protect newborns in high-risk areas during the most vulnerable period of their lives." His team published its findings July 30 in Nature. It noted that the new work hinges on the notion that in the first few weeks of a primate's life -- humans are primates, too -- the body's immune system is naturally more tolerant of "invaders," including gene therapies. The research focused on a tried-and-true form of HIV-fighting gene therapy. It works by programming cells to continuously produce HIV-fighting antibodies. The gene therapy was piggybacked onto a harmless adeno-associated virus (AAV) to help deliver it to the muscle cells of newborn rhesus macaques. Muscle cells were chosen because they are particularly long-lived, Ardeshir's team explained. The gene therapy instructs these cells to produce broadly neutralizing antibodies, or bNAbs, which are capable of neutralizing multiple strains of HIV. It's not the first time bNAbs have been used in gene therapy to fight HIV. However, in prior trials repeat injections were required to keep the immune system vigilant. In the new trial, "we turn these muscle cells -- which are long-lived -- into micro factories that just keep producing these antibodies," Ardeshir explained. When such an approach is used in older monkeys, however, the animals' robust immune systems turn against the therapy, shutting it down. That didn't happen when Ardeshir's team introduced it during a macaque's first few weeks of life. All of the monkeys who got a single shot of bNAbs therapy soon after birth were shielded from infection with HIV for at least three years, with no need for a booster. Tulane researchers said that's roughly the equivalent of a treatment that could ward off HIV in humans deep into adolescence. If the gene therapy was delivered even a bit later -- 8 to 12 weeks after birth -- the young monkey's more developed immune system swung into action to fight it, eroding its effectiveness. Giving the shot soon after birth seemed key, Ardeshir said. "This is a one-and-done treatment that fits the critical time when these mothers with HIV in resource-limited areas are most likely to see a doctor," he noted in a Tulane news release. "As long as the treatment is delivered close to birth, the baby's immune system will accept it and believe it's part of itself." Will it work in human babies? That's not entirely clear, since it's possible infants might be less amenable than monkeys to therapies that are delivered via AAV, the team said. The monkey trial also used only one strain of simian-human immunodeficiency virus, which is similar in some ways to HIV but may not reflect the variety of circulating strains of HIV strains. Still, the research team is hopeful. Giving families a one-shot preventive tool to protect their children would be especially useful in areas where access to repeat medical treatments can be tough, the researchers said. "Nothing like this was possible to achieve even 10 years ago," Ardeshir said. "This was a huge result, and now we have all the ingredients to take on HIV." More information Find out more about HIV and AIDS at the U.S. Centers for Disease Control and Prevention (CDC). SOURCE: Tulane University, news release, July 30, 2025 Copyright © 2025 HealthDay. All rights reserved.

Single Shot Would Protect Children From HIV for Years
Single Shot Would Protect Children From HIV for Years

Newsweek

time5 days ago

  • Newsweek

Single Shot Would Protect Children From HIV for Years

Based on facts, either observed and verified firsthand by the reporter, or reported and verified from knowledgeable sources. Newsweek AI is in beta. Translations may contain inaccuracies—please refer to the original content. Delivering a single shot of gene therapy at birth could offer children protection against HIV for years. A study from the Tulane National Primate Research Center and California National Primate Research Center has identified the first weeks of life—when the immune system is more tolerant—may be the optimal and only window for delivering such gene therapy effectively. "Nearly 300 children are infected with HIV each day," study author and Tulane microbiology professor Amir Ardeshir said in a statement. "This approach could help protect newborns in high-risk areas during the most vulnerable period of their lives." HIV is primarily transmitted to children from the mother breastfeeding after birth. Antiretroviral treatments have shown success in suppressing the virus and limiting transmission (and where the right treatment is available, you can live a normal life with HIV and have an undetectable viral load). However, adherence and accessibility to doctors both decline after childbirth, particularly in areas with more limited health care. "This is a one-and-done treatment that fits the critical time when these mothers with HIV in resource-limited areas are most likely to see a doctor," Ardeshir said. "As long as the treatment is delivered close to birth, the baby's immune system will accept it and believe it's part of itself." A baby receiving an injection. A baby receiving an injection. comzeal/Getty Images In the study, non-human primates received gene therapy that programs cells to continuously produce HIV-fighting antibodies. Timing this right in early life—within the first month—proved critical to the one-time treatment offering long-term protection. "In infants, a single shot at birth kept protective antibody levels high for the entire three-plus years of the study, with no booster required," Ardeshir told Newsweek. "Based on this, we expect protection in people could cover several years as well. "If antibody levels eventually dip, a quick 'top‑up' could be done using a different version of the delivery system to avoid the body's immunity to the first shot, including later in adulthood." However, those in the study treated at 8–12 weeks showed a more developed and less tolerant immune system that didn't accept the treatment as effectively. To deliver the treatment, researchers used an adeno-associated virus (AAV), a harmless virus that can help to deliver genetic code to cells. The virus was sent to muscle cells with a long lifespan and delivered instructions to produce broadly neutralizing antibodies (bNAbs), which are capable of neutralizing multiple strains of HIV. Previous studies found bNAbs to be effective at fighting HIV, but they required repeated infusions, which are costly and come with logistical challenges in low-resource settings. Instead, the researchers turned these muscle cells into "micro factories" to just keep producing the antibodies. A rhesus macaque monkey. A rhesus macaque monkey. Smitt/Getty Images Newborns were found to show greater tolerance and expressed high levels of bNAbs, which successfully prevented infection during simulated breastfeeding and later exposures mimicking sexual transmission. Older infants and juveniles were more likely to have produced anti-drug antibodies that shut down the treatment. Researchers also found that exposing fetuses to the antibodies before birth helped older infants accept the gene therapy later, avoiding the immune rejection that often occurs with age. The researchers believe this type of one-time injection at birth could offer a more cost-effective and feasible real-world solution, while putting less burden on the mother for a follow-up visit. "Although originally designed for babies born to mothers living with HIV, the platform can protect any newborn at substantial risk, including infants whose maternal status is unknown or whose mothers acquire HIV during breastfeeding," Ardeshir said. "In high-prevalence regions, a universal newborn dose could be the most practical public-health approach." Of course, further research will need to consider how the results could translate to human infants and children, who may be less susceptible to AAV-delivered treatments. The study also used one strain of simian-human immunodeficiency virus, which doesn't reflect the variety of HIV strains. However, if successful, it's thought the treatment could dramatically reduce mother-to-child HIV transmission rates in high-risk regions like sub-Saharan Africa, where 90 percent of pediatric HIV cases can be found. "Safety testing and clinical‑grade manufacturing are underway. A first-in-human Phase 1/2 safety trial is being planned and could begin within the next few years. If results look good and regulators agree, we could expand from there," Ardeshir explained. "Importantly, the intramuscular [within the muscle] dose we're using is far lower than the systemic doses now being scrutinized for safety, because we are working with rather than against, the newborn immune system." It may also be possible to adapt the therapy to protect against other infectious diseases like malaria, which disproportionately affects young children in low-income countries. "Nothing like this was possible to achieve even 10 years ago," Ardeshir said. "This was a huge result, and now we have all the ingredients to take on HIV." Do you have a tip on a health story that Newsweek should be covering? Do you have a question about HIV? Let us know via health@ Reference Ardeshir, A., O'Hagan, D., Mehta, I., Shandilya, S., Hopkins, L. L. J., Adamson, L., Kuroda, M. J., Hahn, P. A., da Costa, L. A. B., Fuchs, S. P., Martinez-Navio, J. M., Gardner, M. R., Van Rompay, K. K. A., Magnani, D. M., Lifson, J. D., Gao, G., Farzan, M., Desrosiers, R. C., Das, J., & Martins, M. A. (2025). Determinants of successful AAV-vectored delivery of HIV-1 bNAbs in early life. Nature.

Deaf kids and adults got their hearing back within weeks of having a ‘life-changing' injection
Deaf kids and adults got their hearing back within weeks of having a ‘life-changing' injection

New York Post

time7 days ago

  • New York Post

Deaf kids and adults got their hearing back within weeks of having a ‘life-changing' injection

It's music to our ears. Approximately 11 million people in the US consider themselves deaf or severely hard of hearing. No medications have been approved to treat hereditary hearing loss, but cochlear implants can help people with deafness perceive sound by bypassing damaged parts of the inner ear and directly stimulating the auditory nerve. Advertisement 3 'This is a huge step forward in the genetic treatment of deafness, one that can be life-changing for children and adults,' the study's lead author said. Irina Flamingo – Now, a new gene therapy injection that restored hearing in people with mutations in the OTOF gene offers fresh hope. Researchers injected an AAV vector carrying a healthy copy of the OTOF gene into the inner ear of 10 patients aged 1 to 24. Advertisement The OTOF gene encodes the protein otoferlin, which is essential for transmitting sound signals from the ear to the brain. Without a functional OTOF gene, the critical connection between hair cells in the inner ear and the auditory nerve is disrupted. 3 Researchers injected an AAV vector carrying a healthy copy of the OTOF gene into the inner ear of 10 patients aged 1 to 24. oatautta – Participants began to regain hearing in under a month, with the average perceptible sound threshold dropping from 106 decibels — the level of a lawn mower or motorcycle — to about 52 decibels — normal conversation level — within six months. Advertisement Younger participants fared best, especially those 5 to 8 years old. In one remarkable case, a 7‑year‑old girl regained almost all her hearing and was able to hold a conversation with her mother just four months after treatment. 'This is a huge step forward in the genetic treatment of deafness, one that can be life-changing for children and adults,' Maoli Duan, a consultant and docent at the Department of Clinical Science, Intervention and Technology at the Karolinska Institutet in Sweden, said in a statement. Advertisement 'Smaller studies in China have previously shown positive results in children, but this is the first time that the method has been tested in teenagers and adults, too,' he added. The therapy proved well-tolerated, with no serious side effects reported during the follow‑up period of six to 12 months. The most common reaction was a temporary reduction in neutrophils, a type of white blood cell. 3 Younger participants fared best with the new treatment, especially those 5 to 8 years old. JPC-PROD – 'Hearing was greatly improved in many of the participants, which can have a profound effect on their life quality,' Duan said. 'We will now be following these patients to see how lasting the effect is.' Duan noted that the OTOF gene is 'just the beginning.' 'We and other researchers are expanding our work to other, more common genes that cause deafness, such as GJB2 and TMC1,' he added. 'These are more complicated to treat, but animal studies have so far returned promising results. We are confident that patients with different kinds of genetic deafness will one day be able to receive treatment.'

DOWNLOAD THE APP

Get Started Now: Download the App

Ready to dive into a world of global content with local flavor? Download Daily8 app today from your preferred app store and start exploring.
app-storeplay-store