Xcellbio and Royal Perth Hospital Collaborate on Development of an Automated TIL Manufacturing Workflow

Xcell Biosciences Australia, an affiliate of Xcell Biosciences, and Royal Perth Hospital agreed to collaborate on developing an automated, clinical-scale manufacturing workflow for therapies based on tumor-infiltrating lymphocytes (TILs) using the AVATAR™ Foundry system. The goal is to empower Royal Perth Hospital physicians and clinical researchers to rapidly manufacture TIL therapies for use in clinical trials.

TIL therapies are a promising type of cellular immunotherapy used to treat a broad range of solid malignant tumors, with the first FDA approval of a TIL-based therapy taking place in 2024. These therapies have proven to achieve durable complete responses in patients but require tens of billions of therapeutic cells to achieve sufficient clinical potency.

Current manufacturing processes for TIL therapies are cumbersome and slow, requiring many weeks of manual and labor-intensive efforts to achieve an effective dose, according to Brian Feth, co-founder, and CEO at Xcellbio.

Manufacture potent cell therapies

To address these challenges, Xcellbio has developed the AVATAR platform to expedite the development and manufacture of potent cell therapies. The platform uses a proprietary incubation technology to enhance the metabolic fitness of TILs, resulting in a potent and persistent therapeutic product that can overcome the immunosuppressive tumor microenvironment, explained Feth.

The AVATAR Foundry system was designed to produce cell therapies under current Good Manufacturing Practice (cGMP) protocols in a fully automated capacity.

“At Royal Perth Hospital, we are dedicated to exploring cutting-edge approaches for cell therapies to enable the best care for our patients,” said Zlatibor Velickovic, PhD, director of the Ray and Bill Dobney Cell and Tissue Therapies WA facility at Royal Perth Hospital. “We are excited to team up with scientists at Xcellbio to develop a much-needed automated workflow for TIL manufacturing, and to implement the AVATAR Foundry system as the newest technology innovation in our center.”

Xcellbio scientists will work with the Royal Perth Hospital team to develop an automated TIL manufacturing process based on the AVATAR platform that achieves key goals for cell quality, quantity, and potency. The resulting workflow will be established in the hospital’s cell therapy facility to support clinical trials using TILs.

“The emerging therapeutic class of tumor-infiltrating lymphocytes is incredibly exciting, in particular for solid tumors. Current manufacturing processes require long expansion times and high cell doses to be effective in patients. The AVATAR platform will reduce manufacturing time and increase therapeutic potency,” noted James Lim, co-founder, and CSO at Xcellbio.

“We are pleased to join forces with this group to improve TIL manufacturing and to make these important therapies a more feasible option for patients in Australia and, ultimately, around the world,” said Feth.

Cellares and Cabaletta Bio Complete Manufacturing Technology Adoption Program

Cellares, an Integrated Development and Manufacturing Organization (IDMO), concluded the Technology Adoption Program (TAP) on its automated cell therapy manufacturing Cell Shuttle™ for resecabtagene autoleucel, (rese-cel, previously known as CABA-201).

Cellares’ TAP assessed the feasibility of using the Cell Shuttle platform to automate the manufacturing of Cabaletta’s rese-cel drug product, a CD19-targeting CAR T cell therapy designed to treat patients with a broad range of autoimmune diseases. The TAP successfully delivered automated, concurrent manufacture of multiple rese-cel batches on a single Cell Shuttle, according to Cellares.

Cabaletta Bio and Cellares are now working toward manufacturing cGMP cell therapy batches to be delivered to patients. The Cellares network of global IDMO Smart Factories planned for the United States, Europe, and Japan has the potential to provide Cabaletta Bio with the ability to automate, lower costs, and scale out manufacturing, according to Fabian Gerlinghaus, CEO of Cellares.

Facilitating global expansion

Additionally, Gerlinghaus said, this partnership may facilitate global expansion of rese-cel via rapid technology transfer to additional IDMO Smart Factories. This has the potential to allow Cabaletta Bio to achieve the global scale required to meet the total patient demand across multiple autoimmune diseases, including myositis (~70,000 patients in the United States), scleroderma (~90,000 patients in the United States), and lupus nephritis (~100,000 patients in the United States),  in a fraction of the time and initial investment it would typically take to develop and deliver global supply for these large patient populations, continued Gerlinghaus.

“Through our partnership with Cellares, our teams have successfully achieved proof of concept for the ability to automate the rese-cel cellular drug substance manufacturing process,” said Gwendolyn Binder, president, science and technology at Cabaletta Bio. “We look forward to continuing our work together to complete activities required to enable use of the Cell Shuttle in clinical trials to support the delivery of these potentially curative autologous therapies to more patients with autoimmune diseases.”

“The success of this Technology Adoption Program (TAP) demonstrates the effectiveness of the Cell Shuttle as a scalable, automated, and cost-effective platform for the manufacturing of cell therapies. Working with Cabaletta Bio proves that small biotech companies can successfully partner with Cellares to benefit from next-generation automation,” noted Gerlinghaus.

Engineered yeast boosts D-lactic acid production, advancing eco-friendly biomanufacturing

Great recipes require the perfect combination of ingredients—biotechnology recipes are no exception. Researchers from Osaka Metropolitan University have discovered the ideal genetic “recipe” to turn yeast into a tiny yet powerful eco-friendly factory that converts methanol into D-lactic acid, a key compound used in biodegradable plastics and pharmaceuticals.

This approach could help reduce reliance on petroleum-based processes and contribute to more sustainable chemical production.

The study was published in Biotechnology for Biofuels and Bioproducts.

Lactic acid is widely used in food, cosmetics, pharmaceuticals and bioplastics. It exists in two forms: L-lactic acid and D-lactic acid. Compared to its counterpart, D-lactic acid is much less available and much more expensive.

“Most lactic acid bacteria can only produce L-lactic acid while chemical synthesis methods yield only a mixture of both forms,” said Ryosuke Yamada, an associate professor at Osaka Metropolitan University’s Graduate School of Engineering and lead author of this study.

Seeking a more efficient way to produce D-lactic acid, the team turned to Komagataella phaffii, a yeast capable of utilizing methanol. Their goal was to pinpoint the optimal combination of D-lactate dehydrogenase (D-LDH) genes and promoters in K. phaffii that would maximize the yeast’s ability to produce D-lactic acid from methanol.

D-LDH is a key enzyme responsible for converting precursor molecules into D-lactic acid, while promoters are DNA sequences that regulate gene expression.

After testing five different D-LDH genes and eight promoters, the researchers identified an ideal mix that boosted D-lactic acid production by 1.5 times compared to other methanol-based methods.

“To the best of our knowledge, our engineered yeast achieved the highest-ever reported yield using methanol as the sole carbon source,” Yamada said.

These findings show that engineered yeast strains can be tailored to produce a wide range of useful compounds for commercial use. With growing global concerns over fossil fuel depletion and environmental impact, the ability to synthesize chemicals from renewable carbon sources like methanol is deemed a critical advancement for sustainability.

“This study demonstrates that by carefully optimizing gene and promoter combinations, we can significantly enhance the efficiency of microbial processes, offering a viable alternative to traditional, petroleum-based chemical production,” Yamada said.

Computational drug discovery: Exploring natural products targeting SARS-CoV-2

The COVID-19 pandemic highlighted the urgent need for effective therapeutic agents against SARS-CoV-2. Although vaccines helped control the spread of the virus, the emergence of new variants continues to challenge global health efforts. Small-molecule inhibitors targeting viral proteins could serve as an effective alternative for controlling the spread of COVID-19 at both individual and community levels.

In this vein, a study led by Associate Professor Md. Altaf-Ul-Amin, along with Muhammad Alqaaf and others, explored natural products as potential inhibitors of the SARS-CoV-2 spike protein using computational methods.

As described in their paper published in Scientific Reports, the research team employed molecular docking analysis to screen a diverse library of natural compounds that could act against the spike proteins.

By simulating interactions at the atomic level, they identified several promising molecules with high binding affinities. Such compounds can interfere with viral entry mechanisms, thereby reducing viral activity.

“The variants of spike proteins can be divided into five distinct clusters based on the similarity of their amino acid sequences and functions,” said Associate Professor Md. Altaf-Ul-Amin. “This clustering was performed using an in-house-developed algorithm and software named DPClusSBO, which helped to effectively classify the spike proteins.”

Additionally, the natural products screened in this study were sourced from the KNApSAcK database, which was also developed at the research team’s laboratory. This database contains a comprehensive dataset of natural products useful for drug discovery.

A total of 11 natural compounds were identified: cephaeline, emetine, uzarigenin, linifolin A, caffeine, colchamine, cytidine, (+)-epijasmonic acid, 11-hydroxyvittatine, staurosporin, and paxilline.

Among them, caffeine was particularly noteworthy, as it is commonly found in coffee and caffeinated beverages. Molecular docking along with other analyses revealed that caffeine binds strongly to the spike protein’s active site and exhibits high binding stability. Notably, a drug appropriateness analysis suggested that caffeine possesses excellent solubility and significant potential as an oral drug candidate.

The discovery that a widely known and consumed compound such as caffeine functions as a SARS-CoV-2 inhibitor presents an interesting therapeutic possibility.

In addition to its potential against viral invasion, caffeine plays a dual role in a wide range of medical applications owing to its neuroprotective and anticancer effects. These properties underscore its versatility and demonstrate its potential positive impact on global health.

“Our findings highlight the potential of natural products in the fight against COVID-19. The identified compounds provide a basis for further experimental validation and drug development strategies,” explains Alqaaf, first author of the study.

This research contributes to the broader field of computational drug discovery by demonstrating how bioinformatics and molecular modeling can accelerate the identification of novel viral inhibitors.

Future research will focus on the in vitro and in vivo validation of the identified compounds, as well as potential structural modifications to enhance their antiviral activity.

Gene Therapy Tested in Mice Offers New Hope for People with Dravet Syndrome

Scientists from the Allen Institute and Seattle Children’s Research Institute have announced a breakthrough in the development of gene replacement therapies for Dravet syndrome, a rare form of epilepsy. According to details published in a Science Translational Medicine paper, mice treated with the new therapy survived and had alleviated symptoms and long-term recovery without toxicity and negative side effects. The paper is titled, “Interneuron-specific dual-AAV SCN1A gene replacement corrects epileptic phenotypes in mouse models of Dravet syndrome.”

Dravet syndrome is a severe and difficult to treat condition that is typically caused by a loss-of-function mutation in the SCN1A gene which encodes a sodium channel protein that is involved in brain cell signaling. This mutation leads to problems with interneurons, brain cells that help regulate brain activity. The disease, which is characterized by severe seizures and developmental delays, affects 1 in 15,700 children.

“People who take drugs for epilepsy often complain that the drugs are very impactful, they can slow down the seizures but it changes a lot about their brain,” said Boaz Levi, PhD, associate investigator at the Allen Institute and one of the lead scientists on the study. The goal of the new therapy was “to be very precise” and “just deliver the gene that’s missing.” The result is a treatment that is safer and more effective with significantly fewer side effects, he said.

To ensure that the genes were delivered to the precise location in the defective genes, the researchers used specialized enhancers, short stretches of DNA that act like switches to control when and where specific genes are turned on. They also had to solve the problem of delivering the gene. The conventional approach using adeno-associated viruses would not work for SCN1A because of the gene’s size. Their workaround was to split the gene into two parts and carry each half in separate AAVs. The halves are delivered to the same cells and fused together to make the final gene at their destination.

According to results reported in the paper, dual or single injections of the therapy, dubbed DLX2.0-SCN1A, into mice “did not result in increased mortality, weight loss, or gliosis as measured by immunohistochemistry.”

These results are promising for patients living with Dravet who would “have a severely impacted standard of living” without treatment, Levi said. “We are hopeful this sort of therapy could have a huge impact on families and that’s what’s exciting to me.”

Stomach Lining Mutational Landscape Offers Clues to Gastric Cancer Origins

An international team of scientists has—for the first time, they suggest—systematically analyzed somatic mutations in stomach lining tissue to unpick mutational processes, some of which can lead to cancer. The results from their newly reported study in addition uncovered hints of a potential new cause of stomach cancer that needs further research.

The researchers, including scientists at the Wellcome Sanger Institute, Broad Institute of MIT and Harvard, the University of Hong Kong, and collaborators, sequenced the whole genomes of normal stomach lining samples from people with and without gastric cancer. They discovered that cells with “driver” mutations in cancer genes occupy almost 10% of the gastric lining by age 60 years, and in addition reported the unusual finding that gastric cells in some, but not all, individuals carried three copies of certain chromosomes, hinting toward exposure to an unknown mutagen.

Tim Coorens, PhD, previously at the Wellcome Sanger Institute and now at the Broad Institute of MIT and Harvard, said, “By studying somatic mutations in normal tissues, which we acquire over our lifetimes, we can explore the earliest stages of cancer development. We found that despite constant exposure to acidic stomach contents, the stomach lining is protected. However, in those with gastric cancer, we see higher numbers of mutations in normal cells, resembling the earliest stages of stomach cancer.” The study, Coorens noted, “… adds to a mutation map of the gastrointestinal tract, including the esophagus, stomach, small intestine, and colon, to compare mutation rates and mutational processes across the body.”

The work will support researchers as they explore fundamental mutational processes and compare mutation rates across the body, with a view to further understanding the earliest stages of cancer development. Coorens is first author of the team’s published paper in Nature, titled, “The somatic mutation landscape of normal gastric epithelium,” in which the team concluded: “Our findings provide insights into intrinsic and extrinsic influences on somatic evolution in the gastric epithelium in healthy, precancerous and malignant states.”

Gastric—or stomach—cancer is the fifth most common cancer worldwide, with nearly one million new cases in 2022, the authors reported. It is the third leading cause of cancer-related deaths globally, with the highest number of cases in East Asia and South America.

“The epidemiology of gastric cancer indicates that many extrinsic factors, through exposures and chronic inflammation, influence somatic mutagenesis in the stomach,” they further stated. Factors that increase the risk of developing stomach cancer include being overweight, smoking, and infection with the bacterium, Helicobacter pylori, which can trigger inflammation and stomach ulcers. H. pylori infection causes around 40% of stomach cancers in the U.K.

The stomach acts as a reservoir at the first stage of processing food for digestion, and its contents are acidic. The layer of cells that line the stomach—the gastric epithelium—form gastric glands, or pits, and these contain the cells that can give rise to stomach cancer.

The cells in our body acquire genetic changes, known as somatic mutations, throughout our lifetime. “Over the course of a lifetime, cells in the human body acquire somatic mutations, thus generating genetic diversity and enabling natural selection in tissues,” the team noted. With new DNA sequencing technologies, researchers can now analyze these mutations in normal tissues and trace them back over time, providing insights into aging and the earliest stages of cancer development. “These mutation landscapes provide insights into somatic evolution in normal tissues during an individual’s lifetime and into the earliest stages of cancer development.”

The somatic mutation landscapes of normal epithelial cells lining the esophagus, small intestine, and large intestine have recently been characterized, the authors commented. For their newly reported study, Coorens and colleagues set out to investigate somatic mutations within the gastric epithelium to explore the transition between normal, age-related mutations, and those that go on to form stomach cancer.

The team carried out whole genome sequencing of 238 samples of normal, non-cancerous gastric gland tissue from the stomachs of 30 people from Hong Kong, the United States, and the U.K., of whom 18 had gastric cancer and 12 did not. With laser capture microdissection, they used a laser to precisely dissect individual cells, or glands, from the stomach lining samples for genome sequencing.

The researchers found that despite regular exposure to the acidic contents of the stomach, mutations in normal gastric glands were generated at a similar rate to most cells of the body. This suggests the cells in the gastric epithelium are protected against any toxic effects of the acidic stomach contents.

However, in people with gastric cancer, some of the glands from the normal, non-cancerous stomach lining showed changes under the microscope that resembled the early stages of transitioning to cancer. These normal glands had increased numbers of mutations, which may have contributed to initiating gastric cancer. In the cancerous tissue, the number of mutations was much higher, showing that gastric cancers massively accelerate mutations later during their development.

An unusual finding was that some of the stomach lining cells carried three copies (trisomy) of chromosomes 13, 18, and 20. This has not been seen in other tissues in previous studies, suggesting it is unique to the stomach. Trisomies were found multiple times in some of the individuals, but not present in others. “Remarkably, trisomies were concentrated in a subset of individuals and had often arisen independently and several times in the same individual,” the investigators wrote. This implies these individuals may have been exposed to an unknown, external mutagen. “Our data indicate that rather than a continuous age-associated increase of whole-chromosome duplications, these trisomies were generated at a specific time during the lifespan of each individual and possibly confined to specific regions of the stomach,” the scientists further pointed out.

Co-lead author Suet Yi Leung, MD, at the University of Hong Kong, said: “We discovered an unusual phenomenon, where some individuals had three copies of certain chromosomes—known as trisomy—whereas others did not. We’ve not seen this in any other tissue, and it hints toward an unknown, external mutagen that only some of these people may have been exposed to.”

The scientists also found that by the age of 60 years, “driver” mutations in cancer genes, many of which are known to be mutated in gastric cancer, occupy nearly 10% of the stomach lining. This proportion increases when patients experience severe chronic inflammation, a known risk factor for gastric cancer. “Chronic inflammation can lead to metaplasia, a remodeling of the gastric epithelium to resemble intestinal epithelium, which is thought of as a precursor to overt cancer,” the investigators noted.

As people age, increasing numbers of cells in many of their tissues acquire such driver mutations—genetic changes that directly contribute to the development of cancer. While most cells remain normal, this can lead to abnormal cell growth and division, and can result in cancerous tumors. “The prevalence of mutant clones increases with age to occupy roughly 8% of the gastric epithelial lining by age 60 years and is significantly increased by the presence of severe chronic inflammation,” the authors stated.

This finding points to the need for further research into the mechanism by which chronic inflammation increases the risk of gastric cancer. “Severe chronic inflammation was significantly associated with elevated numbers of driver mutations in gastric glands and overall proportions of mutant epithelium in this study, highlighting a role for chronic inflammation in molding the preneoplastic selection landscape, as also identified in inflammatory bowel,” the team further stated in their paper.

Co-lead author professor Sir Mike Stratton, FMedSci FRS, at the Wellcome Sanger Institute, commented, “Ten years ago, we knew very little about the fundamental processes of mutations that are occurring in our bodies. Now with advanced genome sequencing technologies, we can investigate somatic mutations in all cell types, across various normal tissues. This enables us to look back at the evolution of our cells over a lifetime, to understand the key mutational processes that can lead to cancer. At the Sanger Institute, we are leading the way in investigating the causes and consequences of somatic mutations, and exploring the possibility that somatic mutations may also contribute to diseases other than cancer.”

In an associated News & Views, Callum Oddy and Marnix Jansen, at UCL Cancer Institute, University College London, commented that the study reported by Coorens et al. offers a “… comprehensive analysis that reshapes understanding of how mutations accumulate in normal gastrointestinal tissues and how these changes could set the stage for cancer.” The findings, Oddy and Jansen suggest, “… underscore the value of considering the genetic and epigenetic landscape of normal tissues, not just malignant ones, when studying carcinogenesis.”

“Effective dsRNAs” Combat Cucumber Mosaic Virus in Tests

Researchers at the Martin Luther University Halle-Wittenberg (MLU) have developed RNA-based active agents that appear to reliably protect plants against cucumber mosaic virus (CMV), the most common virus in agriculture and horticulture. Developed to help fight the virus by directing the plant’s natural defenses in the right direction the new molecules, known as efficient double-stranded RNA (edsRNA), demonstrate a broad spectrum effect supporting the plant’s immune system in combating the virus. The team’s laboratory experiments found that 80–100% of treated plants survived CMV infection with a high viral load.

Research lead Sven-Erik Behrens, PhD, at the Institute of Biochemistry and Biotechnology at MLU, and colleagues, reported their findings in Nucleic Acids Research, in a paper titled, “A new level of RNA-based plant protection – dsRNAs designed from functionally characterized siRNAs highly effective against cucumber mosaic virus.” In their paper, the team concluded, “Overall, the results of this study significantly expand the potential for more efficient use of RNA in biological crop protection.”

Virus-induced plant diseases remain a major problem in agriculture that is “recently exacerbated by global trade and climate change,” the authors wrote. Pesticides represent the most common methods for controlling viral infections, but such agents may have non-specific effects on other organisms and can be harmful to humans. “Urgently needed alternative crop protection methods should not only be environmentally sustainable, but also specific, i.e., effective only against a specific target pathogen, and adaptable to the evolution of the pathogen,” the team noted. “RNA-mediated crop protection increasingly becomes a viable alternative to agrochemicals that threaten biodiversity and human health.”

Cucumber mosaic virus is a particularly devastating virus for crops. About 90 species of aphids transmit the virus, which affects more than 1,200 plant species, including numerous agricultural crops such as squash, cucumbers, cereals, and medicinal and aromatic plants. Infected plants are easily identified by a characteristic mosaic pattern on their leaves. Once infected, the plants fail to thrive and their fruits cannot be sold. There are currently no approved agents against CMV.

When a virus infects a plant it uses the plant’s cells as a host. The virus multiplies via its genetic material in the form of RNA molecules in the plant cells. Once injected, these foreign RNA molecules trigger an initial response from the plant’s immune system. Special enzyme scissors recognize and cut the viral RNA molecules. This process produces small interfering RNAs (siRNAs), which spread throughout the plant and trigger a second step of the immune response. The siRNA molecules bind to special protein complexes and guide them to the RNA molecules of the virus. Once there, the proteins begin to break down the harmful RNA molecules of the virus by converting them into harmless, degradable fragments.

“In general, this defense process is not very effective,” said Behrens. “A viral infection produces many different siRNA molecules, but only a few have a protective effect.” Through their newly reported work, the team developed a method to identify functionally effective siRNA molecules, termed esiRNAs, that are highly efficient in the process. “Using an in vitro screen that reliably identifies esiRNAs from siRNA pools, we identified esiRNAs against cucumber mosaic virus …,” the authors explained.

They were able to combine several of these esiRNA molecules into efficient double-stranded RNA molecules (edsRNAs) that could be applied to plants. These edsRNAs act as a package that, soon after entering the plant cells, is broken down into a large number of highly effective siRNA molecules that attack the virus at different sites which can significantly increase the protective effect. “RNA viruses such as the cucumber mosaic virus are dangerous because they can evolve rapidly,” Behrens said. “In addition, the genetic material of this virus is made up of three separate parts, which can get mixed up, further increasing the chance of new mutations. To achieve maximum protection against the virus, our active ingredients target different parts of the genome.”

The team conducted numerous laboratory experiments on the model plant Nicotania benthamiana and was able to show that edsRNA-based active agents reliably protected against cucumber mosaic virus. “The plants in our experiments were infected with a very high viral load: all of our untreated plants died,” reported Behrens. In contrast, 80–100% of the treated plants survived. “… optimal protection was achieved with newly designed multivalent ‘effective dsRNAs’ (edsRNAs), which contain the sequences of several esiRNAs and are preferentially processed into precisely these esiRNAs,” the authors stated. “The esiRNA components can attack one or more target RNAs at different sites, be active in different silencing complexes, and provide cross-protection against different viral variants—important properties for combating rapidly mutating pathogens such as CMV.”

The team has in addition optimized the process of screening for efficient siRNAs and can adapt the procedure to target new viral mutations within two to four weeks. “Time is an important factor: when a new virus variant emerges, we can very quickly modify the active agent accordingly,” Behrens said. The approach may also be applied to other pathogens and pests. “The success of our approach, eNA screen followed by edsRNA design from the identified esiRNAs, promises similar success for other plant pathogens, most of which have significantly less plasticity than CMV,” the authors stated.

Until now, the substances have been administered manually in the laboratory, either by injection or by rubbing them into the plant leaves. The team is working with pharmacist and drug delivery specialist Karsten Mäder, PhD, a professor at MLU to make the RNA-based substances more durable and easier to apply to plants. For example, they could be sprayed on.

At the same time, the researchers are planning field trials to test the RNA-based substances under real conditions. “It will now be important to further test and improve esiRNA and edsRNA actives in combination with suitable formulations in agricultural applications, i.e., greenhouse or field trials,” the investigators wrote in their report. They are also talking to companies about future industrial production. In addition, potential new crop protection products still have to go through an approval process, so it will be some time before a product to combat cucumber mosaic virus enters the market. “However, we are convinced that our approach is feasible. The first crop protection product with an RNA-based active ingredient was recently approved in the USA,” said Behrens.

Ori Biotech Launches Preferred Partner Network

Ori Biotech officials say the company has launched its Preferred Partner Network (PPN), bringing together academic medical centers (AMCs) and contract development and manufacturing organizations (CDMOs) to deliver best-in-class solutions to accelerate the development and commercialization of cell and gene therapies.

The founding members of the Ori Preferred Partner Network in the United States include Charles River Laboratories, CTMC (a joint venture between MD Anderson Cancer Center and Resilience), ElevateBio, Kincell, and other currently undisclosed partners.

PPN membership offers the opportunity to develop core expertise with the new IRO® platform to accelerate cell therapy product development, according to Jason C. Foster, CEO, Ori Biotech.

“By partnering with the top academic institutions and CDMOs globally, Ori is helping to deliver proven solutions that speed time to clinic, reduce comparability risk, and shorten development timelines,” continued Foster. “The PPN enhances the flexibility of therapy developers to choose both best-of-breed technologies and service providers to deliver on their program goals. End-to-end solution providers often try to lock developers in, restricting flexibility, which is the opposite of what we need at this critical juncture for the industry.”

IRO was designed to deliver flexible solutions by integrating with other best-in-breed upstream and downstream technologies providing a streamlined and closed workflow to achieve optimal clinical and commercial success, noted Foster, who added that recently announced technology partnerships, like with Fresenius Kabi, alongside the launch of the PPN, mark a significant step forward in Ori’s mission to enable widespread patient access to life-saving cell and gene therapies.

“Ori’s mission mirrors our own: expedite the delivery of life-saving therapies to patients,” said Matthew Hewitt, PhD, vice president, CTO, manufacturing business division, Charles River.

“CTMC provides comprehensive solutions to our academic and biotech partners, enabling seamless translation from concept to clinic,” explained Jason Bock, CEO, CTMC. “By integrating the automated capabilities of Ori Biotech’s IRO platform, we enhance our technology offerings, streamlining cell therapy manufacturing and accelerating the path to patients.”

“Through the Ori Preferred Partner Network, we are able to offer our partners the necessary technologies and service solutions to accelerate development, manufacturing, and their path to commercialization,” stated Michael Paglia, CTO, ElevateBio BaseCamp.

“As the cell therapy field matures, innovative technologies that simplify the manufacturing of these complex therapies and reduce the cost of goods will enable broad distribution of these life-saving medicines,” pointed out Bruce Thompson, PhD, CTO, Kincell Bio. “Kincell Bio is excited to partner with Ori Biotech on the launch of the IRO platform to help our partners and clients ensure their cell therapy products reach patients in need.”

Dogs’ noses decoded: Optical sensor unveils canine brain’s olfactory prowess

A pioneering study investigating the brain activity of dogs during scent detection has unveiled crucial insights into their remarkable olfactory capabilities. Researchers at Bar-Ilan University have developed an optical sensor capable of remote sensing dogs’ brain activity in three key regions—the olfactory bulb, hippocampus, and amygdala—that play a critical role in how dogs distinguish between different smells. This breakthrough could lead to the development of a compact, non-invasive device capable of interpreting and translating a dog’s olfactory perceptions for human understanding.

In the study published in the Journal of Biophotonics, scientists employed a cutting-edge detection structure system using laser technology and a high-resolution camera to capture brain activity in real-time from four dog breeds.

These dogs were exposed to four distinct scent stimuli—garlic, menthol, alcohol, and marijuana. The data were then analyzed using a machine-learning algorithm revealing that the amygdala plays a significant role in scent differentiation, highlighting the emotional and memory-related aspects of odor processing.

“The findings show that the amygdala is crucial in the way dogs process and react to odors, with specific scents triggering distinct emotional and memory responses, and we are capable of optically detecting their brain activity in this region,” said Prof. Zeev Zalevsky, from the Kofkin Faculty of Engineering at Bar-Ilan University. “This discovery could be the first step toward creating a device that enables us to better understand and interpret the unique way dogs perceive and differentiate smells.”

The study introduces an innovative method of brain activity analysis through laser-based speckle pattern detection, a remote, non-invasive technique that has never been applied to canine brain activity. Unlike traditional methods such as fMRI or EEG, this approach allows researchers to observe brain responses without requiring the dog to be sedated or confined to bulky equipment. This opens up new possibilities for studying dogs in real-world environments, making the technique both affordable and accessible for further research.

Dogs have long been celebrated for their exceptional sense of smell, and this research further illuminates the advanced processes that occur in their brains when detecting odors. With an olfactory system far more developed than humans, dogs can detect a broader range of odors, with specialized receptors in their noses that allow them to process and distinguish even the faintest scents.

This new research offers a glimpse into the intricate workings of the canine brain as it processes different smells, presenting a promising avenue for future applications in areas such as drug detection, medical diagnostics, and search-and-rescue missions.

“Our next step is to develop a portable, Wi-Fi-controlled device equipped with a mini camera and laser system, which could be mounted on a dog’s head and used to monitor its olfactory responses in real time,” said Dr. Yafim Beiderman from Prof. Zalevsky’s Optical Research Lab at Bar-Ilan University.

“This could significantly enhance the way dogs are used in scent detection, from detecting illegal substances to diagnosing diseases in humans, all while deepening our understanding of how they perceive the world around them. More importantly, this real-time sensing could bypass the need to train dogs to utilize their scent abilities.”

The implications of this research could also revolutionize the way dogs are utilized in law enforcement, health care, and beyond. As dogs continue to be invaluable partners in scent detection, this device could provide a means of translating their highly specialized abilities into data that is useful for humans, fostering a stronger connection between the two species.

Current Antivirals, Including Tamiflu, Ineffective Against H5N1 Virus in Cows’ Milk

H5N1 influenza virus continues to circulate in cattle (the virus has been found in dairy cows’ milk and has infected farm workers) causing public health officials to consider preparations for a potential outbreak (or even pandemic) in humans. One aspect of pre-pandemic planning is testing currently approved antivirals against influenza A viruses (like H5N1) circulating in peridomestic species.

In a new study, results suggest that in a preclinical model, two FDA-approved flu antivirals did not successfully treat severe H5N1 infections. Additionally, the researchers found that the route of infection, whether through the eye, the nose, or the mouth, significantly impacts a treatment’s effectiveness.

The findings are published in Nature Microbiology in the paper, “Baloxavir improves disease outcomes in mice after intranasal or ocular infection with Influenza A virus H5N1-contaminated cow’s milk.”

“Our evidence suggests that it is likely going to be hard to treat people severely infected with this bovine H5N1 bird flu strain,” said Richard Webby, PhD, St. Jude Department of Host-Microbe Interactions. “Instead, reducing infection risk by not drinking raw milk and reducing dairy farm workers’ exposures, for example, may be the most effective interventions.”

Though H5N1 infections in people have been rare, there are more than 60 people to date who have become infected from dairy exposures in the current outbreak. Some were infected through exposure to contaminated raw cows’ milk, such as dairy workers who were infected through splashes or aerosolized particles reaching their noses or eyes. Given the risks to human health, the scientists used a mouse model to test how each antiviral drug worked against the virus when it was obtained through three different exposure routes.

“In general, baloxavir [Xofluza] caused a greater reduction in viral levels than oseltamivir [Tamiflu], but neither was always effective,” said Jeremy Jones, PhD, St. Jude Department of Host-Microbe Interactions.

The researchers studied exposure routes that included the eye, mouth, and nose, which are the most common ways to become infected with the virus. The oral route, which mimics drinking raw infected cow’s milk, caused the worst infections that were hardest to treat.

“The virus spread orally far beyond its normal infection of the lungs,” Webby said. “It expanded to the brain and the bloodstream, and the antivirals failed to stop it or improve survival outcomes.”

In contrast, findings showed that baloxavir controlled infections through the eye fairly well. These results are particularly relevant as the ocular route appears to be the common infection pathway for people who work directly with dairy cows. “Baloxavir conveyed 100% survival compared to 25% with oseltamivir,” Jones said. “So, we are seeing enhanced benefits from baloxavir for the ocular infection route.”

Results were mixed for the nasal route. Baloxavir reduced viral levels better than oseltamivir, but the virus still reached the brain. Both antivirals increased survival, with baloxavir and oseltamivir achieving a 75% and 50% survival rate, respectively.

“We showed our existing antivirals’ effectiveness against H5N1 bird flu is route and drug dependent, in some cases doing almost nothing,” Webby said. “Therefore, while we explore different drug combinations and doses, we need to do anything we can to reduce the risk of infection, as that is the best way to protect people from this virus right now.”

error: Content is protected !!