SESSION IV: I WISH I KNEW… (PRECLINICAL)

SESSION IV. I WISH I KNEW... (PRECLINICAL) CHAIR: Leticia Toledo-Sherman, PhD—Rainwater Charitable Foundation
Challenges in HTT Lowering Biomarker Development for Huntington’s Disease –
A Case Study
Douglas Macdonald, PhD—CHDI Foundation
Alzheimer’s Disease Drug Discovery: NMEs vs. NCEs, Pros and Cons of EachAllen Reitz, PhD—ALS Biopharma
Challenges in Drug Development for Neurodegenerative Disease/ATM Kinase Inhibitors for Huntington’s Disease, a Case StudyLeticia Toledo-Sherman, PhD—Rainwater Charitable Foundation
Development of Novel Stress Kinase Inhibitor D. Martin Watterson, PhD—Northwestern University

Session Overview

Leticia Toledo-Sherman, PhD - Chair, Session IV

Dr. Leticia Toledo-Sherman is Senior Director of Drug Discovery for the Tau Consortium and Adjunct Assistant Professor of Neurology at UCLA. In her role at the Foundation, she provides strategic direction and oversight of the drug discovery portfolio. Dr. Toledo-Sherman brings critical expertise and best practices in neurodegenerative disease drug discovery to accelerate the development of therapies for Tauopathies.

Previously, she was Director of Medicinal Chemistry and Computer-Aided Drug Design at the CHDI Foundation, leading drug discovery programs for therapeutic development in Huntington’s Disease (HD). Prior to joining CHDI, Dr. Toledo-Sherman was Executive Director of Chemistry at Lymphosign (Pharmascience Inc), leading the development of therapeutics for blood cancers. From 2000 to 2004, she led a multi-site, multidisciplinary team using chemical proteomics and bioinformatics to discover therapeutic targets and to investigate the mechanism of action of drugs. Dr. Toledo-Sherman started her professional career as a leading scientist at Kinetix Pharmaceuticals (acquired by Amgen), where she implemented an in silico platform aimed at rational drug design of kinase inhibitors targeting multiple therapeutic areas. She has served on the Science Advisory Board of the SGC-UNC Kinase Unit and is currently on the Science Advisory Board of The Chemical Probes Portal.

Dr. Toledo-Sherman is an advocate and supporter of open science at the pre-competitive drug discovery stage. She received a PhD in Organic Chemistry from The State University of New York at Stony Brook and did postdoctoral research at the Massachusetts Institute of Technology and The Skaggs Institute for Chemical Biology at The Scripps Research Institute in La Jolla, California.

Q&A

How can I join the Tau Consortium?

The Tau Consortium is a consortium put together by its Scientific Advisory Board (SAB), based on interest of the Principal investigator (you) and publications and the research priorities the Consortium’s SAB has deemed most important to advancing the field of Tauopathy research. The main areas being funded are Basic Discovery Biology (understanding the underlying biology of tauopathies and the creation of tools and models), Drug Discovery and Translation (discovery and development of therapeutics for tauopathies, from target discovery to end stage preclinical development) and Clinical research and trials (natural history studies, patient registries, interventional clinical trials. An important focus for the Tau Consortium is the development of Biomarkers and PET imaging agents. You can learn more about these efforts at https://tauconsortium.org/research/.

Can you tell us more about the funding opportunities for young investigators, PhD students and postdocs at Rainwater Charitable foundation?

Besides the funding opportunities as described in question 1, which is by invitation only, the Rainwater Foundation runs co-funding initiatives

Challenges in HTT Lowering Biomarker Development for Huntington’s Disease – A Case Study

Douglas Macdonald, PhD—CHDI Foundation

Douglas Macdonald, PhD

Dr. Douglas Macdonald is a molecular neuropharmacologist with extensive drug discovery and development experience in the pharmaceutical and biotech industries. At CHDI he forges and manages external partnerships, coordinates in-licensing and out-licensing opportunities, and works with the Foundation’s biology and chemistry groups to advance therapeutic programs across the portfolio of scientific and research projects. His scientific focus is on the development of HTT lowering therapies (gene silencing and gene therapy) as well as biomarkers and HTT protein quantitation assays.

Prior to CHDI, Dr. Macdonald was a Senior Research Scientist in the CNS Molecular and Functional Neuropharmacology group at Sanofi-Aventis (Bridgewater, NJ) where he led several R&D programs for neuropsychiatric disorders. Before that, he conducted research in the CNS Department at the Schering-Plough Research Institute (Kenilworth, NJ) and in the Merrifield laboratory at the Rockefeller University (New York, NY). He received his PhD in Pharmacology and Experimental Therapeutics from Boston University School of Medicine, his MS in Medical Sciences from the Department of Biomolecular Medicine at Boston University, and his BS in Chemistry from Trinity College (Hartford, CT).

He is currently a member of Boston University School of Medicine’s Dean’s Advisory Board, a member of the Board of Fellows at Trinity College, a member of the NIH/NINDS Neuropharmacology and Diagnostics Small Business Innovation Research Study Section, serves on the SAB for the CureDuchenne and an advisor to the ALS Association, and is a Lecturer at the University of Southern California School of Pharmacy and Regulatory Sciences.

Q&A

Considering that ASOs have been around for a long time, why do you think that they gain popularity only in the recent past?
DM: I would say that it’s due to the technological advances in the field, such as novel ASO chemistries which can afford a longer duration of action as well as a better understanding of delivery methods, especially to the CNS, and distribution of the ASO to specific regions of interest.
Did you find sex difference in CSF mHTT?
DM: None to date, but the data set remains relatively small and we will be looking at that and others possible differentiating factors.
Thank you for your very interesting presentation. How do I get in contact with CHDI? Are all the collaboration CHDI-initiated?

DM: The best way is to go to our website (https://chdifoundation.org/) and review the “Community Resources” section and reach out to the listed contact for your area of interest. You may also contact me at any time.

Do you think that IONIS would have been able to successfully complete their trials, and potentially commercialize their drug, without a biomarker? In other word, how critical is the development of your biomarker?
DM: In my opinion, biomarkers are essential and can enable, and even accelerate, compound advancement in the clinic. There is a large attrition rate in drug development and the use of biomarkers is one way to mitigate risk.
Hi Douglas, great talk! In one of your last slide, you showed that there was a multi-teams efforts. Can you comment on IP ownership?
DM: In multi-team programs IP ownership will depend on the particular partnership. CHDI works with academic researchers and institutions, contract resource organizations, biotechnology companies, as well as large pharmaceutical companies. As you can imaging each one of these partnerships will have different levels of IP ownership.
It took about 15 years since the beginning of your collaboration with IONIS. Knowing all the things that you know today, what is the one single thing that you would do differently, and that would allow you to save a significant amount of time? Thank you
DM: That is a great question and it is always important to look retrospectively to see what lessons have been learned. In this case, the Huntington’s ASO program was extremely novel. At the time, IONIS had just started working in the CNS space and along the way we learned a lot about how different ASO chemistries can afford longer durations of action and perhaps most importantly, the team developed a better understanding of direct CNS delivery methods and the relationship to the distribution of the ASO to specific regions of interest. I suggest you contact team members from IONIS for more information regarding these areas. But furthermore, recall that at the start of the program we could not measure mHTT in HD patient CSF, and that was a significant technological advance that enabled the progression of the program to where it is today. One would always like to go faster, but sometime technology needs to catch up.

Alzheimer’s Disease Drug Discovery: NMEs vs. NCEs, Pros and Cons of Each

Allen Reitz, PhD—ALS Biopharma

Allen Reitz, PhD

Dr. Allen Reitz has had more than 38 years of experience in the pharmaceutical industry, including nearly 26 years with Johnson & Johnson. He led the Central Nervous System Medicinal Chemistry research effort including for Alzheimer’s Disease for 16 years at the Spring House, PA facility of JNJ. He has invented eight compounds that have entered human clinical trials, including mazapertine and troriluzole, the later of which is currently in Phase II/III clinical trials for the treatment of Alzheimer’s Disease (www.t2protect.org). He has over 160 scientific publications and 68 issued U.S. patents, is on the Editorial Advisory Board of ACS Med. Chem. Lett. and edits the journal Current Topics in Medicinal Chemistry. He has extensive experience in project and portfolio management, target validation, hit triage, hit to lead and lead optimization medicinal chemistry, eADME profiling, and preclinical candidate selection.

He is also Adjunct Professor at Drexel University College of Medicine and holds an Executive Masters in Technology Management from the University of Pennsylvania (Wharton, Penn Engineering). Dr. Reitz founded and is CEO of both Fox Chase Chemical Diversity Center, Inc. and ALS Biopharma, LLC since 2008.

Q&A

Thank you Allen, you made it sounds so simple, this was a fantastic presentation! Can you elaborate more on the first generation screening? Which assay did you use?
AR: First generation screening for prodrugs was PK analysis, and this is typically the case for prodrugs, with the thought being that if you can achieve the desired PK you will have a viable therapeutic effect, which you can also demonstrate in secondary screening. In the case of prodrugs, we sought to be stable in the GI track and in liver microsomes with instability in plasma, across multiple species including human, mouse, rat and dog. The general screening paradigm is described in McDonnell, M. E.; Vera, M. D.; Blass, B. E.; Pelletier, J. C.; King, R. C.; Fernandez-Metzler, C.; Smith, G. R.; Wrobel, J.; Chen, S.; Reitz, A. B. Riluzole prodrugs for melanoma and ALS: design, synthesis, and in vitro metabolic profiling. 2012, Bioorg. Med. Chem., 20, 5642-5648. For TDP-43, our primary assay is an alpha-screen looking at the ability of compounds to bind to TDP-43, presumably monomeric TDP-43, and prevent the binding of nucleic acid substrate, as described in Cassel, J. A.; Blass, B. E.; Reitz, A. B.; Pawlyk, A. C. Development of a Nonradiometric Assay for Nucleic Acid Binding to TDP-43 Suitable for High-Throughput Screening Using AlphaScreen® Technology. J. Biomol. Screening, 2010, 15, 1099-1106.
Why did you synthetized dipeptide conjugates? And why did you add a third amino acid?
AR: Many (but not all) of the single amino acid analogs underwent an internal 5-membered ring spirocyclization reaction of the alpha-amine onto the 2-carbon of the benzimidazole as described in Pelletier, J. C.; Velvadapu, V.; McDonnell, M. E.; Wrobel, J. E.; Reitz, A. B. Intramolecular rearrangement of a-amino acid amide derivatives of 2-aminobenzothiazoles. Tetrahedron Lett. 2014, 55, 4193-4195, so we added a second amino acid which we reasoned could cleave either with proteases or cyclization to a diketopiperazine. The problem there was that our top dipeptide lead had unacceptable hERG, so we added another amino acid which got rid of hERG and also had the benefit of being actively transported by the PepT1 transporter which helps to increase oral bioavailability and minimize the variability of exposure when eating or fasting.
Can you clarify why a third aminoacid was added to create troriluozole?
AR: See answer to the above question.
If esterification is a very common approach in drug discovery, is it also common to extend the IP to related esters?
AR: As I noted in the talk, most esters in drugs or drug candidates are prodrugs followed by esterase cleavage to give the pharmaceutically active agent. However, some esters are relatively metabolically stable and are the pharmaceutically active agent on their own without being cleaved. Yes, of course, IP should be extended to include esters, even prophetically – that is, prodrugs of active agents can be described in a patent application such as by the formation and cleavage of esters.
Isn’t the patient the ultimate customer? Can you provide some additional information, or even recommend reading or provide links, regarding customer identification?
AR: That’s an interesting question – “isn’t the patient the ultimate customer”. First, a book to recommend: Steve Blank and Bob Dorf, The Startup Owner’s Manual, 2012, K&S Ranch, Inc. The patient is the ultimate end-user, but usually not the ultimate customer. The physicians who prescribe and the formularies and insurance companies that include or exclude drugs from their lists are more often than not the ultimate customers, even if the end users are the patients. It’s a bit of a semantic puzzle, but worth thinking about, and I do recommend the book listed above.
Amazing presentation, thank you for sharing! For a very early startup, how can you have a balanced portfolio of risk-reward?
AR: There’s no boilerplate answer on how to have a balanced portfolio of risk-reward. Complicating this is the often a project that seems low risk turns out to be harder than first anticipated, and vice versa. The only operational way to have a balanced portfolio is to create one – that is, have a many bets approach even within a single project, in which risk is mitigated with “Plans Bs” and defined pivot and walk-away points. Experience and intuition play over-sized roles in drug discovery as well.

Drug Discovery at the Tau Consortium; ATM Kinase Inhibitors as POC agents for Huntington’s Disease, a Case Study

Leticia Toledo-Sherman, PhD—Rainwater Charitable Foundation

Leticia Toledo-Sherman, PhD - Chair, Session IV

Dr. Leticia Toledo-Sherman is Senior Director of Drug Discovery for the Tau Consortium and Adjunct Assistant Professor of Neurology at UCLA. In her role at the Foundation, she provides strategic direction and oversight of the drug discovery portfolio. Dr. Toledo-Sherman brings critical expertise and best practices in neurodegenerative disease drug discovery to accelerate the development of therapies for Tauopathies.

Previously, she was Director of Medicinal Chemistry and Computer-Aided Drug Design at the CHDI Foundation, leading drug discovery programs for therapeutic development in Huntington’s Disease (HD). Prior to joining CHDI, Dr. Toledo-Sherman was Executive Director of Chemistry at Lymphosign (Pharmascience Inc), leading the development of therapeutics for blood cancers. From 2000 to 2004, she led a multi-site, multidisciplinary team using chemical proteomics and bioinformatics to discover therapeutic targets and to investigate the mechanism of action of drugs. Dr. Toledo-Sherman started her professional career as a leading scientist at Kinetix Pharmaceuticals (acquired by Amgen), where she implemented an in silico platform aimed at rational drug design of kinase inhibitors targeting multiple therapeutic areas. She has served on the Science Advisory Board of the SGC-UNC Kinase Unit and is currently on the Science Advisory Board of The Chemical Probes Portal.

Dr. Toledo-Sherman is an advocate and supporter of open science at the pre-competitive drug discovery stage. She received a PhD in Organic Chemistry from The State University of New York at Stony Brook and did postdoctoral research at the Massachusetts Institute of Technology and The Skaggs Institute for Chemical Biology at The Scripps Research Institute in La Jolla, California.

Q&A

Do you know if ATM has been implicated to other neurodegenerative diseases?

There is an emerging body of literature of the role of ATM in neurodegenerative diseases; see following reviews and references within.

  • Burn et al. P. Cytoplasmic ATM protein kinase: an emerging therapeutic target for diabetes, cancer and neuronal degeneration. Drug Discov Today. 2011;16(7-8):332‐338.
  • Fielder, Edward, von Zglinicki, Thomas, and Jurk, Diana. ‘The DNA Damage Response in Neurons: Die by Apoptosis or Survive in a Senescence-Like State?’ 1 Jan. 2017 : S107 –S131.
  • Choy KR, Watters DJ. Neurodegeneration in ataxia-telangiectasia: Multiple roles of ATM kinase in cellular homeostasis. Dev Dyn. 2018;247(1):33‐46. doi:10.1002/dvdy.24522
If the mechanism of action is not fully understood (e.g. in your case the underlying biology of ATM in DNA repair) would you say that is not worth it to investigate a specific target?

It is a bit of a chicken and an egg problem. One of the reasons we set out to create brain penetrant selective ATM inhibitors was partly to further the understanding of the underlying biology of ATM in DNA repair in the context of Huntington’s disease. While there are other tools, siRNAs, ASOs, antibodies, CRISPR etc. a sharp small molecule chemical tool that can be delivered to the right tissue and site of action is a very useful tool to interrogate biology. Tools
like siRNA, ASO, antibodies and CRISPR have poor brain penetration. We already had indication that ATM is involved in HD biology/pathology from GWAS, SiRNA knockdown, and the cross of the ATM Knockdown mice with the HD mice which rescue several landmark phenotypes of HD in those mice, but being able to dose a small molecule that can inhibit the ATM at the site of action and follow these effects in a controlled fashion was very informative to a program (not all the data was shown given time constrains in the presentation, we have a new manuscript in preparation). We felt this was key, before further resources are deployed to generate a clinical candidate. One of the goals of CHDI is to de-risk targets by creating brain penetrating sharp probes for those targets. Chemical Biology with sharp chemical probes has been very effective in advancing therapeutics.

In your last slide you showed that small tweaks can overall lead to strong additive effects. How much time and effort should be spent on?

This is one of the most underappreciated areas of medicinal chemistry and compounds optimization and where chemists often go for big changes in potency (>100fold) and often discard the opportunities that a 10 fold-here and a 10 fold-there tweaks can offer when combined. In this program we encountered different philosophical perspectives, but at the end with a few precedents the team was convinced it was working. I would advise keeping your eyes open for these opportunities, specially, when you have a rational design strategy and the SAR is solid. However, this can only happen if your data is very tight and your assays highly reproducible, to assure that these small effects are real and not just a variability of the assay). I would not say to go only for small effects but be aware of the opportunities they present.

Can you tell us more how to include and consider co-morbidities in drug development or clinical trials?

See literature below for a perspective of the challenges comorbidies present in AD.

Kahle-Wrobleski K, Fillit H, Kurlander J, Reed C, Belger M. Methodological challenges in assessing the impact of comorbidities on costs in Alzheimer's disease clinical trials. Eur J Health Econ. 2015;16(9):995‐1004. doi:10.1007/s10198-014-0648-7 and
https://alzheimersnewstoday.com/2018/11/14/comorbidities-common-disabling-in-alzheimers-and-dementia-patients-uk-study-finds/

Can you clarify what do you mean with “proper PK”?

Yes, I should have defined that better. By proper PK, I meant to dose compounds via a route IV/PO (oral) that allows to understand how much compound got absorbed, distributed, was metabolized and was excreted by determining all pharmacokinetics parameters, (avoid dosing compound in drinking water for example, where it is hard to understand how much compound was up taken by the animal). You should collect sufficient time point to derive a good area under
the curve and understand compounds half-life and elimination. For brain penetration, it is important to understand the free fraction in brain and free compound concentrations to assure you have obtained compound exposures that cover or are above the effective concentrations (EC50) of your compounds at eliciting its effect at the target or mechanism in cells. So if your compound has an EC50 of say 50nM, free brain concentrations of 50nM or above will be needed
to assure any effect observable in vivo can be attributed to the compound. I recommend reading this helpful review and perspective on brain PK ( (Li Di, Demystifying Brain Penetration in Central Nervous System Drug Discovery,J. Med. Chem. 2013, 56, 1, 2–12https://pubs.acs.org/doi/10.1021/jm301297f)

What should I do if I don’t know my molecular target, but I have good efficacy in vitro and in vivo data?

While it is difficult to assert an efficacy without understanding compound exposure at the target site and a pharmacodynamic effect, there are techniques that can help you deconvolute your compound mechanism of action. It is important to assess, however if your compounds is present in sufficient quantities to engage that target of mechanism so you can attribute an efficacy.

There are various chemical biology and proteomics techniques that allow you identify the target of a compound identified in a phenotypic assay:

  • Activity-based protein profile (ABPP)
  • Cellular thermal shift assay (CETSA)
  • Compound-immobilized beads
  • Photoaffinity labeling (PAL

Here are some good reviews that describe these techniques:

https://www.nature.com/articles/nrd2410/
https://pubmed.ncbi.nlm.nih.gov/30392561/
https://linkinghub.elsevier.com/retrieve/pii/S1570963918301249

There are several CROs that offer target deconvolution services.

Case Study from an Academic Consortium: Development of Novel Stress Kinase Inhibitor Drug Candidate

D. Martin Watterson, PhD—Northwestern University

D. Martin Watterson, PhD

Dr. Martin Watterson serves in an advisory role to pharmaceutical and biotechnology companies in the areas of process and risk analysis. In addition to industry consulting, Dr. Watterson serves on advisory boards for small business start-ups, biotechnology companies, and non-profit organizations in the area of CNS drug discovery and development. His personal CNS drug development experience includes the discovery and preclinical development of novel small molecule therapeutic candidates that attenuate disease related to synaptic dysfunction, as well as participation in development of protein replacement therapeutics.

Dr. Watterson is the G.D. Searle Endowed Chair Professorship at Northwestern University, where he is also Professor of Pharmacology in the Feinberg School of Medicine. Previous relevant activities at Northwestern include the founding of an academic drug discovery research and training program characterized by the generation of multiple CNS drug candidates taken into preclinical and clinical development through the leveraged use of Foundation and NIH funding. He also served in various administrative positions, including Department Chair, University Center Director, and Curriculum Co-Director.

Prior to Northwestern, he held faculty positions at The Rockefeller University, where he was an Andrew Mellon Fellow, and at Vanderbilt University Medical Center, where he was Professor of Pharmacology and an Investigator in the Howard Hughes Medical Institute. Dr. Watterson is the recipient of the 2016 Melvin R. Goodes Prize recognizing researchers working in promising areas of drug discovery for Alzheimer’s disease and related dementias.

Q&A

Could you recommend additional reading about the recursive CNS drug discovery platform?

Directly related to the case study example covered in the talk, I would recommend Roy et al., (2019), Journal of Medicinal Chemistry, 62:5298-5311, and the references cited therein.

If your drug will not be efficacious in AD, do you think that would be worth it to test in other neurodegenerative disease? Which one and why?

Yes. Based on documented biological rationale in the peer reviewed literature, the most proximal alternative clinical indications would be tauopathies (Roy et al., 2019, JMC 62:5298) and certain Autism Spectrum Disorders (Robson et al., 2018, PNAS, 23,115:E10245). The rationale is based on pathophysiology progression mechanisms in the clinical diseases and the evidence of MW150 target engagement and efficacy in relevant preclinical animal models. MW150 is essentially a phase 2 ready clinical therapeutic candidate at this time, so the preclinical GxP data package and phase 1 clinical trial suggests these if funding becomes available. Because the preclinical development was focused on common pathophysiology progression mechanisms, other diseases where those mechanisms are contributors to disease susceptibility or progression are reasonable considerations for the future.

Great talk, I really leaned a lot, thank you very much. Approximately, how long it took you to develop MW150? If you knew how to avoid elapsed time gaps, how much time could you have saved?

Once the basic research on CNS protein kinases identified how to circumvent the challenges in the field, it took a bit over five years to take a lead compound to a candidate ready for start of phase 1a clinical trial by the spin-out biotech. If we had made better use of complementary foundation and NIH funding to minimize the cycle of submission/peer review/award date time gaps, we most likely would have removed 18 months or more from the timeline.

Can you tell us more about non-productive alternative approaches? Any recommendation to avoid to fall in love with an idea? Does it help to have clear go-no-go decision points?

Even if one has clear go-no-go decision points in advance, the larger challenge is taking a new molecule into disease areas where there are no disease modifying therapeutic precedents. Therefore, a less linear path to the goal is the starting point. A number of alleys must be pursued in try/fail probing – a bit like mapping an unknown territory. The overall Go/NoGo decision is based on a set of activity outcomes from multi-disciplinary studies, not a single test or milestone. Once an experimental protocol in a particular discipline (med chem; pharmacology; animal models) is established and results are being produced readily, the temptation is to continue generating more data in that disciplinary silo in the absence of strategic input from other disciplines. How far down an exploratory alleyway one goes before hitting a dead end, or “wall” as it called in med chem, is the tough decision. For example, medicinal chemistry refinement can hit experimental “sweet spots” where diversification of a precursor allows rapid generation of an entire family of analogs to test in a facile and quantitative primary assay. There is a temptation to fall in love with an emerging beautiful SAR series if one stays only within that disciplinary silo. The key activity is to pick landmark structures at strategic points to test in selected secondary pharmacology assays. The secondary pharmacology outcomes can support a mid-campaign decision to stop or shelve the seductive SAR path that is falling into place. One would then re-visit what alternative chemical space to explore that is a better fit with the campaign’s long term goal. This is where a recursive platform is especially useful as one gains greater knowledge in each round of passage with less expenditure of time and effort. Using the case study presented and examples in the Roy et al. (2019) report, the sole pursuit of higher affinity (lower IC50 values) for the primary target in the absence of selected secondary pharmacology screens would have delivered a different series of drug candidates. One set would have been higher affinity inhibitors with off-target activity in the kinome and non-kinome target space. Another set would have been high affinity inhibitors in an in vitro assay, but the deliverable would have been a ligand with metabolic liabilities or less attractive bioavailability. The adage is to focus on drug candidates, not beautiful ligands. For example, the addition to every compound’s evaluation at a critical point was liver microsome stability and screens for a troublesome cross-over kinase target (Roy et al., 2019). The addition of key secondary pharmacology in vitro screens at strategic end points in the campaign yielded a candidate with higher potential for selectivity that was partially de-risked for pharmacological properties. Addition of focused pilot pharmacodynamic and in vivo pharmacokinetic screens at a later step moved the campaign towards it primary goal, a de-risked drug candidate to take into phase 1 clinical development. The recursive platform using strategic secondary pharmacology inputs decreased the amount of time and effort spent on non-productive synthetic chemistry and later stage in vivo animal studies. The overall Go/NoGo decision points based on well-considered milestones were not changed throughout this process.

Can you elaborate on your last point, that is leverage infrastructure and knowledge base from previous campaigns?

There are two aspects: the literature and the intellectual/physical infrastructure of the inter-disciplinary team. For the former, the prevailing view in the literature at the start of our campaign contributed to the decision to bias our primary efforts away from amyloid pathway effects and add increased early testing of safety pharmacology and toxicology de-risking. In this way, we had higher probability to have a drug candidate delivered to first-in-human studies that was both a potential alternative approach as well as a complementary approach amenable to a clinical multi-drug environment for a complex disease. In terms of the intellectual and physical infrastructure of the inter-disciplinary team, the presence of a disease focused structural genomics infrastructure and the availability of qualified assays to probe rapidly neuroinflammation and synaptic dysfunction pathophysiology mechanisms were key infrastructure strengths among team members. The prior experience with azine synthetic chemistry and pharmacology was also leveraged in order to generate initial structures with pharmacological potential for early testing and design of a preclinical drug development campaign.

Can you tell us the most difficult challenge you faced when you started Neurokine, as well as the most rewarding thing that happened so far?

The most difficult challenge, for me personally, in assisting Neurokine Therapeutics (NKT) get their start was the process of getting the campaign out of academia and into the real world where viable products are generated in a regulated industry. The most rewarding thing so far is the cooperative efforts of NKT members and academic colleagues to get the clinical asset into phase 1 first-in-human safety studies and to generate a commercial scale GMP clinical drug supply for start of first-in-patient trials when funding becomes available. The time line using a virtual spin-out and peer-reviewed funding is remarkable to me.