“I am grateful that thoughts don’t appear above our head” – Unknown

“Be the reason someone smiles today.” – Unknown

Remember to send your nominations for quarterly awards to Lauren by March 19th. Winners to be awarded during the quarterly meeting on March 24th. Recognize your team members for their outstanding work!

“Listen and silent are spelled with the same letters. Think about it.” -unknown

“Whatever you must do today….Do it with the confidence of a 4-year-old in a Batman cape” -Unknown

“Happiness is letting go of what your life is supposed to look like and celebrating it for everything that it is.” – Mandy Hale

“The time is always right to do what is right”. -Martin Luther King, Jr.

“You are amazing. Remember that.” – Unknown

“A mentor is not someone who walks ahead of us to show us how they did it. A mentor walks alongside us to show us what we can do.” – Simon Sinek

“May your dreams be bigger than your fears. May your actions speak louder than words. May your faith in yourself be stronger than your emotions.” – Annie Larson

Vedere Bio was founded in June 2019 with $21 million series A funding and was acquired by Novartis in a lucrative deal valued at up to $280 million ($150 million upfront)1 18 months later. This is exceptional even in the fast-paced M&A world and one contributing factor may be that Vedere’s technology has the potential to be a game changer in gene therapy.  Vedere’s novel optogenetics platform could be used to develop novel gene therapies for retinal disorders like age-related macular degeneration (AMD) and retinitis pigmentosa that can lead to blindness. It’s well known that cataracts and retinal eye diseases are the most common causes of permanent blindness, and while cataracts are treated surgically, retinal eye diseases typically do not have a standard treatment regimen. In developed countries, retinal diseases are the most common cause of irreversible blindness and AMD is the most common retinal eye disease in older people. AMD does have a few therapeutic options such as laser photocoagulation and anti-angiogenic therapies (for wet AMD) but there is an unmet clinical need for long-term therapies and the more prevalent dry form of AMD has very limited therapeutic options. Gene therapies are being actively investigated but one of the complications is the fact that there are several therapeutic targets for retinal disease – more than 250 different genetic mutations have been reported for retinitis pigmentosa alone. Gene therapy that targets a specific disease driver gene would be beneficial only to the patients with that mutation, thus limiting the addressable patient population.


Vedere Bio’s technology is unique in that it does not depend on the presence of specific genetic mutations and can address a broader patient population. Some retinal diseases like AMD and retinitis pigmentosa can cause widespread death of photoreceptor cells (rods and cones) resulting in vision loss. Vedere’s strategy is to target retinal cells that are not destroyed during the disease process. The technology was originally developed at UC Berkeley in the labs of Ehud Isacoff and John Flannery and focuses on the development of adeno-associated virus (AAV) vectors that express light sensing opsin proteins and can be directly injected into the vitreous space. The goal of this approach is to reverse blindness by using an AAV expressing green cone opsin targeted to retinal ganglion cells in the inner retina. Normally, these cells are not sensitive to light but the presence of the green cone opsin protein makes them light sensitive, so they are able to function as surrogates for photoreceptor cells and generate electrical signals for the brain to interpret as vision.

Another unique feature is the AAV vector that was engineered to specifically infect cells in the inner retina. The AAV serotype 2 viruses with tissue specific opsin expression infect the retinal ganglion cells in the inner retina– the tissue specific expression simplifies the delivery method as the virus can be directly injected into the vitreous space instead of the subretinal area, which is a more complicated process. The use of the green cone opsin is a technological innovation as optogenetic methods have used microbial opsins that have high response rates but need a strong light stimulus which could damage the retina. Conversely, rhodopsin and melanopsin from retinal ganglion cells are sensitive but have a slow response to light. The medium wavelength green cone opsin solves both challenges as it is sensitive to dim light and has a fast response rate. An added benefit is that the green cone opsin protein allows visual adaptation in normal light for 3D object visualization.

This opens up the possibility that people with advanced retinal disease may be able to see again and regain a better quality of life so Vedere’s innovative technology could be a true game changer in ocular gene therapy.

Interested in reading more about the development and testing of the AAV in the rd1 mouse model of blindness? Check out the original paper from March 2019.

1 https://www.prnewswire.com/news-releases/novartis-acquires-vedere-bio-a-novel-optogenetics-aav-gene-therapy-company-301162269.html

NorCal Spring Symposium

May 7, 2024

Genentech
455 East Grand Avenue
Building 44
South San Francisco, CA

A_Rafie

Meet Amir.
Catch up with him during the NorCal Spring Symposium or click here to contact him directly by e-mail.

Amir Rafie,
Associate Director, Business Development

Association for Research in Vision & Ophthalmology (ARVO)

May 5-9, 2024

Seattle, WA

A_Hall

Meet April.
She will be attending ARVO with several of our JOINN colleagues from China. You can catch up with her in the exhibit area (booth #4803), or click here to contact her directly by e-mail.

April Hall,
Chief Business Offer

Unlocking the Mysteries of Drug Development Using Novel Tools and Modalities
Meeting followed by a happy hour (35 Gatehouse Drive, Waltham)

April 25, 2024
Waltham, MA

Hosted by AstraZeneca
35 Gatehouse Drive
Waltham, MA
Meeting followed by happy hour (35 Gatehouse Drive, Waltham)

G_Gagnon

Meet Gavin.
Catch up with Gavin during BAPTG or click here to contact him directly by e-mail.

Gavin Gagnon,
Manager, Business Development

OTSS Hosted Webinar
SOT Ocular Toxicology Specialty Section (OTSS)

April 24, 2024
10:00 AM to 12:00 Noon (US EST, UTC -5)

General Considerations of Using Non-Human Primates in Non-Clinical Ocular Drug Research
Speaker: Eric (Wankun) Xie, MD, PhD

BIONNOVA
Beijing Innovation Forum 2024

April 24-25, 2024

Beijing, China

Chinese

Neurodegenerative diseases encompass several disorders that are typically associated with the death of specific neuron types resulting in loss of motor, cognitive and other abilities, and most of these diseases are ultimately fatal. It is estimated that there are more than 600 neurodegenerative disorders that impact 50 million Americans each year1. Despite the growing unmet clinical needs, there has been limited progress in developing new therapies for neurodegenerative diseases. Drug discovery and development rely heavily on preclinical in vitro and in vivo models to study disease biology, identify new drug targets and test therapies. While different models have distinct advantages and drawbacks, the selection of a given model should be carefully done to answer specific biological questions. For example, cell-based models derived from primary disease state neurons or induced pluripotent stem cells (iPSC) lines are widely used to study disease pathology and identify mechanism of action for new drug targets2. Additionally, cell-based models are well suited to screen small molecules or biomolecules to identify candidate therapies to test in animal models. Traditionally, cell-based models consisted of simple 2D cell cultures but increasingly there is a shift towards using more complex 3D cell models that are cultured on a scaffold to mimic the tissue environment2. While the in vitro models are useful in the early stages of drug discovery, the gold standard to evaluate therapeutic efficacy and safety are animal models.

Several rodent models of neurodegenerative disease are used in preclinical discovery programs for new therapies. These models include transgenic models where specific disease-causing mutations are introduced or chemically induced models to induce neurological damage3. Despite the large number of publications and funding, mouse models of neurodegenerative disease have been shown to have limited translation to human patients as therapies that were shown to be efficacious in mouse models had very limited effect in humans3. One of the key reasons for this is that the rodent CNS is very different from the human CNS in terms of anatomy, physiology and neuronal complexity3. Additionally, rodents have a short lifespan so it is difficult to model age-related neurodegenerative disorders such as Alzheimer’s disease and Parkinson’s disease. Nonhuman primates (NHPs) are better suited to model neurodegenerative diseases as the NHP CNS is well suited to evaluate changes in cognition, brain function and motor skills that are hallmarks of neurodegenerative diseases. Aged NHPs have been reported to develop age-related issues such as neuron loss, plaque formation and cognitive deficits similar to humans4. However, it is expensive and complicated to maintain an NHP colony for long time periods to study the natural development of neurodegenerative diseases. Therefore, the development of induced disease models is of interest. For example, the injection of beta-amyloid containing brain tissues into NHP brains were shown to induce plaque formation, neuroinflammation and other neuronal issues4 associated with Alzheimer’s disease. However, NHP models of Alzheimer’s disease have not been widely adopted likely due to ethical and cost issues. Animal models of Parkinson’s disease can be induced by the injection of specific chemicals such as MPTP or 6-hydroxydopamine. These compounds can be injected directly into the brain or systemically into the vasculature, skin or muscle. NHP models injected with MPTP recapitulated the motor skill deficits associated with Parkinson’s disease4. More recently, intracerebral injections of gene therapy vectors encoding mutant alpha-synuclein or Lewy body extracts in rhesus macaques and cynomolgus NHPs induced neuronal cell loss and increased expression of alpha-synuclein but these physiologic changes did not translate to motor skill changes4.

While Alzheimer’s disease and Parkinson’s disease are challenging to model since the exact genetic drivers of disease development are not fully known. In contrast, the disease driver for Huntington’s disease has been identified as an expansion in the number of CAG repeats in the huntingtin gene. Researchers have introduced the mutant huntingtin gene using lentiviral vectors into specific brain regions of cynomolgus macaques and demonstrated the development of Huntington’s disease symptoms4. An attempt to develop a transgenic NHP model was reported but the animals with the mutant huntingtin gene had very short lifespans4. To overcome this disease-related challenge, researchers have developed iPS cell lines from transgenic NHP models of Huntington’s disease to study disease biology and reported the development of an NHP iPSC-derived astrocyte model for Huntington’s disease5.

In summary, while NHP models are highly translational and recapitulate several hallmarks of neurodegenerative diseases, the complexity of developing and maintaining these models pose significant challenges.

References:

1https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1280411/

2https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9063566/

3https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6615039/

4https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5886328/

5https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6428250/