Impact case study database
- Submitting institution
- University of Dundee
- Unit of assessment
- 5 - Biological Sciences
- Summary impact type
- Technological
- Is this case study continued from a case study submitted in 2014?
- No
1. Summary of the impact
Parkinson’s disease affects 6 million people worldwide . One genetic cause is mutation in the LRRK2 gene. Research of Professor Dario Alessi and colleagues has: (1) underpinned over USD1billion investment in the development and clinical trial of LRRK2 inhibitors as therapeutics to treat Parkinson’s; (2) provided ‘gold standard’ assays and tools to monitor LRRK2 activity that R&D in global pharma has relied on; (3) identified LRRK2-related biomarkers that are critical to evaluating the effectiveness of therapeutics in clinical trials; and (4) produced tools/reagents for LRRK2 preclinical research that are being marketed worldwide and used by industry.
2. Underpinning research
Parkinson’s Disease is a debilitating progressive disorder of the nervous system. Genetic breakthroughs have uncovered genes linked to inherited forms of Parkinson’s. One cause of familial Parkinson’s is mutation within a protein kinase gene called LRRK2 (for leucine- rich repeat kinase 2). When this became apparent in 2004, little was known of the physiological function of LRRK2. Research led by Alessi revealed robust quantitative evidence that the most common pathogenic mutation, located within the kinase domain of LRRK2, increases its kinase activity [R1]. This was an early strong indication that hyper-activity of LRRK2 could be the mechanism by which mutations in this protein cause disease and indicated that drug inhibitors of LRRK2 activity might offer a potential route to treatment.
To facilitate research on LRRK2, Alessi developed LRRKtide, a synthetic LRRK2 substrate for assessing LRRK2 kinase activity [R1]. He also discovered key phosphorylation sites on LRRK2 that mediate its biological interactions and raised monoclonal antibodies against them [R2]. These have been universally used in academia and industry to assess in vivo efficacy of LRRK2 inhibitors. In 2011, in partnership with collaborator Nathanael Gray (Harvard), Alessi published the creation of the first potent and selective inhibitor of LRRK2 [R3].
Progress in the research field was stymied for a number of years by a lack of understanding of the implications of increased LRRK2 kinase activity on cell biology. Alessi devoted 12 years to identifying physiological substrates of LRRK2. In 2016 the breakthrough came from a collaboration between Alessi and Matthias Mann (Max Planck Institute), Merck, GlaxoSmithKline (GSK) and the Michael Fox Foundation for Parkinson’s research (MJFF) with the seminal discovery that proteins known to be involved in trafficking between cell compartments (members of the Rab family of small GTPases) were the validated physiological phosphorylation targets of LRRK2 [R4]. All known Parkinson’s-associated LRRK2 mutations significantly enhance Rab phosphorylation in vivo and this disrupts their normal trafficking within the cell [R5].
In order to analyse the signalling pathway, the Alessi lab developed a panel of sensitive, phospho-specific monoclonal antibodies to monitor the activity of the endogenous LRRK2 pathway in vivo [R5]. This significant undertaking resulted in a major step change in the biochemical research tools available to easily measure the activity of the LRRK2 pathway. Subsequently, Dr Esther Sammler (UoA1) together with Alessi used these reagents to develop the first biomarker assay measuring LRRK2 phosphorylation of pRab10 in human blood cells [R6]. Alessi and Sammler also discovered a link between LRRK2 and how another Parkinson’s mutation causes disease, indicating a value for LRRK2 inhibitors even in patients without LRRK2 mutation.
3. References to the research
[R1] Jaleel, M, Nichols, RJ, Deak, M, Campbell, DG, Gillardon, F, Knebel, A & Alessi, DR (2007) LRRK2 phosphorylates moesin at threonine-558: characterization of how Parkinson's disease mutants affect kinase activity, Biochemical Journal, vol. 405, no. 2, pp. 307-317. DOI: 10.1042/BJ20070209
[R2] Nichols, RJ, Dzamko, N, Morrice, NA, Campbell, DG, Deak, M, Ordureau, A, Macartney, T, Tong, Y, Shen, J, Prescott, AR & Alessi, DR (2010) 14-3-3 binding to LRRK2 is disrupted by multiple Parkinson's disease-associated mutations and regulates cytoplasmic localization, Biochemical Journal, vol. 430, no. 3, pp. 393-404. DOI: 10.1042/BJ20100483
[R3] Deng, X, Dzamko, N, Prescott, A, Davies, P, Liu, Q, Yang, Q, Lee, J-D, Patricelli, MP, Nomanbhoy, TK, Alessi, DR & Gray, NS (2011) Characterization of a selective inhibitor of the Parkinson's disease kinase LRRK2, Nature Chemical Biology, vol. 7, no. 4, pp. 203-205. DOI: 10.1038/NCHEMBIO.538
[R4] Steger, M, Tonelli, F, Ito, G, Davies, P, Trost, M, Vetter, M, Wachter, S, Lorentzen, E, Duddy, G, Wilson, S, Baptista, MAS, Fiske, BK, Fell, MJ, Morrow, JA, Reith, AD, Alessi, DR & Mann, M (2016) Phosphoproteomics reveals that Parkinson’s disease kinase LRRK2 regulates a subset of Rab GTPases, eLife, vol. 5, e12813, pp. 1-28. DOI: 10.7554/eLife.12813
[R5] Lis, P, Burel, S, Steger, M, Mann, M, Brown, F, Diez, F, Tonelli, F, Holton, JL, Winglok Ho, P, Ho, S-L, Chou, M-Y, Polinski, NK, Martinez, TN, Davies, P & Alessi, DR (2018) Development of phospho-specific Rab protein antibodies to monitor in vivo activity of the LRRK2 Parkinson’s disease kinase, Biochemical Journal, vol. 475, no. 1, pp. 1-22. DOI: 10.1042/BCJ20170802
[R6] Fan, Y, Howden, AJM, Sarhan, AR, Lis, P, Ito, G, Martinez, TN, Brockmann, K, Gasser, T, Alessi, DR & Sammler, E (2018) Interrogating Parkinson's disease LRRK2 kinase pathway activity by assessing Rab10 phosphorylation in human neutrophils, Biochemical Journal, vol. 475, no. 1, pp. 23-44. DOI: 10.1042/BCJ20170803
Key research grants relevant to this case study:
Alessi, DR. Understanding signalling pathways mutated in inherited disorders. Medical Research Council. (2013-present). Value: GBP6,415,000.
Alessi, DR. Regulation of the Parkinson's LRRK2 protein kinase by Rab29. Parkinson’s UK. (2018‐2021). Value: GBP91,389.
Alessi, DR. Towards a unifying theory of Parkinson's disease: Investigation of the biochemical and genetic role of Rab GTPases (2016-2018) Medical Research Council. Value: GBP240,758.
Alessi, DR. Characterisation of the LRRK2 protein kinase, mutated in inherited Parkinson’s disease. Medical Research Council. (2008-2011). GBP254,088
Alessi, DR. The Michael J. Fox Foundation for Parkinson’s Research. Cumulative grants from 2012- 2021. GBP 1,795,583
Sammler, E. Biomarkers in Parkinson's disease: A blood test to measure LRRK2. Parkinson’s UK. 2017-2019. Value: GBP 49,270
Sammler, E. Bench-to-bedside Parkinson’s disease research: Biomarkers in LRRK2 Research proposal. NHS Research Scotland (NRS) and Scottish universities. (2019-2023). Value: GBP1,046,335
4. Details of the impact
Beneficiaries:
Pharmaceutical industry
Patients
Biotech reagent companies
Impacts:
- Focussing and enabling commercial drug discovery on LRRK2 inhibitors
The robust evidence that a common LRRK2 pathogenic mutation in Parkinson’s increased its kinase activity, and the development of a synthetic LRRK2 substrate, had critical impact in directly stimulating the development of LRRK2 inhibitor therapeutics by industry [E1]. The Vice President of MJFF and former Merck LRRK2 Inhibitor Program Biology Lead confirms Alessi’s contribution:
“During the early days of LRRK2 kinase inhibitor programs, all pharmaceutical companies relied on his published work to establish primary assays to screen for LRRK2 kinase inhibitors…[and used his]… synthetic LRRK2 substrate…to screen their compound libraries…These discoveries…had a major impact on the field since they stimulated the pharmaceutical industry to embark on LRRK2 kinase inhibitor drug discovery and development…[His tools] …directly facilitated the entry of the first LRRK2 kinase inhibitors into the clinic in 2017. People with Parkinson’s are now testing these potential therapies thanks to Dr. Alessi’s efforts” [E1].
- Underpinning investment in LRRK2 inhibitors and their entry into the clinic
As a result of the progress facilitated by Alessi’s research, biopharma company Denali Therapeutics developed two brain-penetrant LRRK2 inhibitors that have generated positive results in Phase 1 and Phase 1b clinical trials in healthy volunteers and Parkinson’s patients [E2]. One Denali inhibitor is now in preparation for late-phase clinical trials. This success stimulated significant investment in the company in 2020, with Biogen announcing a USD1billion deal to co-develop and co-commercialise Denali’s LRRK2 inhibitors [E3]. Denali document many direct impacts of the research in enabling their trials [E3] which led to economic and patient benefits.
Global pharmaceutical company GlaxoSmithKline (GSK) has directly collaborated with Alessi on LRRK2 since 2008. GSK published nine World Intellectual Property Organization LRRK2 inhibitor patents 2015-2019 that cite Alessi’s 2007 research [R1]; six also cite the 2016 paper [R3] which they had involvement with [E4]. In 2018, GSK announced a $300M partnership with DNA testing company 23andMe to expedite recruitment to their LRRK2 inhibitor clinical trials by targeting individuals with LRRK2 mutations [E4]. Worldwide, more than 43 patents were published 2014-2019 on small molecule LRRK2 inhibitors [E5]. Many belong to global pharmaceutical companies such as GSK, Pfizer, Merck, Roche and Genentech who all cite Alessi’s work [E5].
- Adoption of LRRK2 biomarkers with direct impact in enabling clinical trials
Alessi’s research discoveries provided the two blood biomarkers that are essential to evaluating the effectiveness of treatments for LRRK2-mutation Parkinson’s in clinical trials. Monitoring, via Alessi’s UDD2 antibody, phosphorylation at a key site in LRRK2 became “ a gold standard measure of target engagement for LRRK2 kinase inhibitors” according to MJFF [E1]. MJFF’s precompetitive alliance with Denali, Pfizer, Merck and Biogen, the ‘LRRK2 Detection in PBMC Consortium’, depended on UDD2 to develop a biomarker for their therapeutic trials [E6]. Similarly, referring to Alessi’s identification of Rab GTPases as LRRK2 substrates, Denali say they “ immediately recognized the importance of this published insight and applied this to…[develop] high throughput clinical grade assays to measure Rabs as a pathway engagement biomarker” [E3]. Both types of biomarker were measured in participant/patient blood during the Denali clinical trials to evaluate LRRK2 inhibitor effectiveness [E2]. Alessi’s research also enabled Denali to find the correct inhibitor dose for the trials [E3]. MJFF and others are sponsoring clinical studies in the USA and Europe measuring the same blood biomarkers to see if they can identify people with enhanced LRRK2 pathway activity whether or not they have LRRK2 mutations or Parkinson’s symptoms [E7]. The Vice President MJFF states:
“All of the target engagement work being conducted by pharmaceutical companies to measure LRRK2 and Rabs in…[blood] cells is due to Dr. Alessi’s efforts and collaboration with MJFF to generate and make available at cost, crucial monoclonal antibodies” [E1].
- Marketing of LRRK2 products by laboratory reagent supply companies
LRRK2-related reagents from the Alessi lab are marketed to industry and academia by Abcam, NeuroMab, and the Dundee MRC-PPU Reagents and Services facility [E8]. Revenue from sales to industry by MRC-PPU was approximately GBP200,000 over the REF period [E8] and is likely to have been higher for the commercial vendors. These reagents are used extensively by Pharma, including Pfizer, Boehringer Ingelheim and Merck, for Parkinson’s preclinical research [E9].
- Bringing hope and encouraging people with Parkinson’s to participate in research
In 2017, Parkinson’s UK established the Dundee Parkinson’s Research Interest Group (DRIG). Researchers from the University of Dundee regularly interact with DRIG, enabling people affected by Parkinson’s to input to the research. The Chairperson of DRIG comments: “ DRIG has benefitted greatly from our association with the MRC PPU…[We contribute] …through occasionally commenting on research proposals…as well as by funding some small research projects.” [E10]. The group keeps up-to-date with Sammler’s genetic testing for early-onset or inherited Parkinson’s patients diagnosed in Scotland. People identified as having LRRK2 mutations become eligible to join clinical studies she leads for Dundee/UK such as the industry-led Rostock International Parkinson’s Disease Study enrolling participants worldwide [E7], and the LRRK2 International Parkinson’s Disease Study [E7]. Informed involvement in such research benefits people with Parkinson’s:
“…working with the MRC-PPU gives us not just hope, but also knowledge and a little bit of control over our future, which strengthens our determination to carry on and ultimately beat Parkinson’s” – Chair of DRIG [E10].
5. Sources to corroborate the impact
[E1] Testimonial from the Vice President, The Michael J Fox Foundation for Parkinson’s Research.
[E2] Phase 1/1b Clinical trials of LRRK2 inhibitors DNL201 and DNL15 using measurement of biomarkers identified by Alessi, pS935 and pRab10, as secondary outcome measures: ( NCT04551534) 16th September 2020; ( NCT03710707) 18th October 2018; ( NCT04557800) 22nd September 2020; ( NCT04056689) 14th August 2019.
[E3] Testimonial from Chief Medical Officer and Head of Development, Denali Therapeutics, and related press release.
[E4] GSK World Intellectual Property Organisation LRRK2 patent list citing R1 and R4 and GSK Press Release.
[E5] Xiao Ding & Feng Ren (2020) ‘Leucine-rich repeat kinase 2 inhibitors: a patent review (2014-present)’, Expert Opinion on Therapeutic Patents, vol. 30, no.4, pp275-286. DOI: 10.1080/13543776.2020.1729354 and example patents from GSK (WO2019012093A1), Pfizer (US9695171B2), Roche/Genentech (WO2013139882A1) and Merck (US9493440B2).
[E6] Publication by MJFF of results of pre-competitive collaboration with industry partners with the goal of optimizing the measurement of pLRRK2 in human PBMCs. Alessi is named as advisor and use of his UDD2 antibody supplied by Abcam is noted in Table 1.
[E7] Clinical Observational studies to assess LRRK2 activity (including phosphorylated LRRK2 and LRRK2 phosphorylated Rabs, detected using Alessi’s antibodies) in blood cells as biomarkers in Parkinson’s Disease: (NCT03545425) 4th June 2018; (NCT03866603) 7th March 2019; (NCT04214509) 2nd Jan 2020; (NCT04436848) 18th June 2020.
[E8] Panel of LRRK2 associated reagents developed by Alessi marketed by (a) Abcam (Anti-LRRK2 antibodies ab133450, ab133518) (b) anti-LRK2 clone 8G10 from NeuroMAb (75-308) and (c) LRRK2 reagents available to purchase from the UoD MRC PPU Reagents and Services website, and statement corroborating sales of reagents.
[E9] Biopharma publications corroborating use of Dundee UDD2 antibody (Abcam ab133450) in industry LRRK2 inhibitor programmes.
Pfizer LRRK2 assay: Galatsis, P et al. (2014), ‘Kinase domain inhibition of leucine rich repeat kinase 2 (LRRK2) using a [1,2,4]triazolo[4,3-b]pyridazine scaffold’, Bioorganic & Medicinal Chemistry Letters vol. 24, no. 17, pp 4132-40. DOI: 10.1016/j.bmcl.2014.07.052
Merck LRRK2 assay along with LRRKtide and antibodies against it replicated and marketed by ThermoFisher: Fell, MJ et al. (2015) ‘MLi-2, a Potent, Selective, and Centrally Active Compound for Exploring the Therapeutic Potential and Safety of LRRK2 Kinase Inhibition’, Journal of Pharmacology and Experimental Therapeutics. vol. 355, no. 3, pp.397-409. DOI: 10.1124/jpet.115.227587
Boehringer Ingelheim: Speidel, A et al (2016) ‘Leucine-Rich Repeat Kinase 2 Influences Fate Decision of Human Monocytes Differentiated from Induced Pluripotent Stem Cells’, PloS One vol. 11,11 e0165949. DOI: 10.1371/journal.pone.0165949
[E10] Testimonial from the Chair of the Dundee Parkinson’s Research Interest Group.
- Submitting institution
- University of Dundee
- Unit of assessment
- 5 - Biological Sciences
- Summary impact type
- Technological
- Is this case study continued from a case study submitted in 2014?
- No
1. Summary of the impact
In 2012, the WHO declared its intent to eliminate Human African Trypanosomiasis (HAT) through a ‘test-and-treat’ strategy, requiring new screening tools that are accurate, easy to use and can be deployed at all levels of the health system. Ferguson’s team identified a protein from the trypanosome surface (Invariant Surface Glycoprotein-65 or ISG65) as a superior diagnostic antigen for HAT. This directly led to the development of a new Rapid Diagnostic Test for HAT incorporating this antigen. The test has been adopted by the global not-for-profit Foundation for Innovative New Diagnostics as their preferred test for HAT screening. Further, in anticipation of post-elimination surveillance, they are using ISG65 in a combined test for HAT screening and malaria diagnosis.
2. Underpinning research
Human African Trypanosomiasis (HAT), or sleeping sickness, is caused by subspecies of the parasite Trypanosoma brucei transmitted by infected tsetse flies. It is generally fatal if untreated. Although the prevalence of HAT decreased dramatically over the past 20 years due to strict control measures, history has shown that it can quickly re-establish to epidemic proportions if surveillance is relaxed. It is endemic in 36 sub-Saharan African countries with 57 million people at risk. The WHO has targeted HAT for elimination of community transmission by 2030. Accurate testing to identify infected individuals is critical to enable timely treatment and for active surveillance to identify reemergence in areas of previous disease elimination. When Ferguson’s team began the research, HAT screening used the relatively primitive CATT (Card Agglutination Test for Trypanosomiasis) test where trypanosomes fixed to a card are used to test patient’s serum for the presence of antibodies. The CATT needs a refrigerated ‘cold-chain’ during distribution, and trained personnel with electrical lab equipment for patient testing. It has severe limitations for large-scale use for community screening in remote locations.
Ferguson recognised that new approaches were needed. In research published in 2013 [R1], he identified improved diagnostic antigens and a more suitable platform for inexpensive field-based testing. Taking an unbiased approach, he used antibodies from the blood of HAT patients to probe a mixture of trypanosome proteins and see which proteins they bound to. This identified a trypanosome surface protein, the Invariant Surface Glycoprotein ISG65, as the best diagnostic antigen. Ferguson obtained experimental tools from Prof Mark Carrington at the University of Cambridge and subsequently produced ISG65 in an engineered bacterium (recombinant ISG65) ensuring unlimited future supply without the need to grow parasites. To progress this research towards a usable diagnostic, Ferguson and colleagues developed a prototype lateral flow Rapid Diagnostic Test (RDT) in collaboration with Dundee-based immunoassay development and manufacturing company BBI Solutions. The performance of this recombinant antigen prototype RDT [R1], and a subsequent version incorporating a second native antigen also discovered in the Ferguson lab [R2], fulfilled the desired characteristics of a priority diagnostic test for FIND, the Foundation for Innovative New Diagnostics, a global not-for-profit product development partnership accelerating the development of diagnostic tests for poverty-related diseases.
Prior to Ferguson’s involvement, the first-generation RTD commissioned by FIND for HAT used native antigens purified from live parasites. This has several disadvantages; few laboratories have the specialized facilities and expertise needed to grow the parasites and purify the antigens, and batch-to-batch quality issues can interrupt production of tests. WHO and FIND therefore required second-generation RTDs to be developed using recombinant antigens [R1]. At the suggestion of Ferguson, FIND commissioned an independent (blinded) three-way side-by- side comparison of the existing dual native antigen test (SD BIOLINE HAT), a dual recombinant prototype of the same antigens, and the Dundee dual antigen prototype. Statistical analysis of the data showed that one of the two best recombinant diagnostic antigens was Dundee’s ISG65 [R3]. All of Dundee’s knowhow and reagents were transferred free of charge to FIND who contracted Abbott (Standard Diagnostics Inc, South Korea, hereafter SD Inc) to manufacture a second-generation RDT (BIOLINE HAT 2.0) for front-line use in Africa with recombinant ISG65 as one of its two diagnostic antigens. The new kit is cheap, can be stored at room temperature, is easy to use even in remote resource-poor settings and will give rapid results in 15 minutes from a pin-prick of blood.
3. References to the research
[R1] Sullivan, L, Wall, SJ, Carrington, M & **Ferguson, MAJ (**2013) 'Proteomic selection of immunodiagnostic antigens for human African trypanosomiasis and generation of a prototype lateral flow immunodiagnostic device', PLoS Neglected Tropical Diseases, vol. 7, no. 2, e2087.
DOI: 10.1371/journal.pntd.0002087
[R2] Sullivan, L, Fleming, J, Sastry, L, Mehlert, A, Wall, SJ & **Ferguson, MAJ (**2014) 'Identification of sVSG117 as an immunodiagnostic antigen and evaluation of a dual-antigen lateral flow test for the diagnosis of human african trypanosomiasis', PLoS Neglected Tropical Diseases, vol. 8, no. 7, pp. e2976. DOI: 10.1371/journal.pntd.0002976
[R3] Sternberg, JM, Gierlinski, M, Biéler, S, Ferguson, MAJ & Ndung'u, JM (2014) 'Evaluation of the diagnostic accuracy of prototype rapid tests for human African trypanosomiasis', PLoS Neglected Tropical Diseases, vol. 8, no. 12, pp. e3373. DOI: 10.1371/journal.pntd.0003373
Key research grants relevant to this case study:
Ferguson, MAJ. The biosynthesis of glycoproteins in Trypanosoma brucei: basic and translational research. Wellcome. (2008-2013). Value: GBP1,707,314.
Ferguson, MAJ. Protein glycosylation in trypanosomes: defining and exploiting a biological system. Wellcome (2013-2020). Value: GBP2,360,250.
4. Details of the impact
Impacts:
Partnership with FIND and Abbott to accelerate HAT diagnostic test deployment
Development of improved, more cost-effective diagnostic, BIOLINE HAT 2.0
Successful clinical study of diagnostic BIOLINE HAT 2.0
Adoption and implementation of the new, supply chain-resilient HAT diagnostic
Commissioning of a combined RDT prototype for both HAT and malaria
- Partnership with FIND and Abbott (SD Inc) to accelerate HAT test deployment
Ferguson identified FIND as the ideal partner to help translate his research into a product for clinical use. In 2014, FIND and the University of Dundee entered a formal partnership, signing a
License and Material Transfer Agreement under which reagents developed by Ferguson were made available free of charge for the development of an improved RDT for HAT [E1]. FIND provided the bridge to the private sector and commissioned Abbott (SD Inc) to produce BIOLINE HAT 2.0 using Dundee’s recombinant ISG65. Dundee provided the ISG65 expression clone, optimised protocols, and consultancy advice to enable scale-up of rISG65 production by Abbott [E2]. Both FIND and Abbott benefited from the partnership with Dundee. Abbott benefit through for-profit production of the RDT, while FIND Senior Project Manager says [E2]:
“FIND is grateful for Professor Ferguson's continued engagement and assistance in technology transfer to Standard Diagnostics and Abbott… we acknowledge the University of Dundee's significant research contributions to achieving FIND's goals in supporting the elimination of HAT. ”
- Development of improved, more cost-effective rapid diagnostic test BIOLINE HAT 2.0
By providing antigen ISG65 and prototype technology, Dundee initiated a workflow at FIND that included evaluations on affordability, performance, market analysis, and risk assessment [E2]. FIND Senior Project Manager describes their conclusions and confirms that Dundee research:
“had a significant impact, prompting FIND’s development of their second-generation ‘all- recombinant’ RDT for HAT… The removal of reliance on the supply of native antigens, which can be laborious and dangerous to produce, provided the opportunity for a step change in the robustness of HAT screening…The all-recombinant nature of this second-generation device is critical to RDT production resilience, quality and cost control. The Bioline HAT 2.0 is affordable at US$ 0.50/test and stable for up to 2 years at temperatures up to 40°C… Packaging of HAT 2.0 will be smaller… giving distribution cost savings. The RDT format also provides several advantages over the traditional CATT test, including no requirements for electrical supply, ancillary equipment, a cold-chain or highly-trained operators. All of these factors are important for deploying HAT screening programmes in sub-Saharan Africa” [E2].
- Successful clinical study of rapid diagnostic test BIOLINE HAT 2.0
In 2015, FIND began a multi-centre clinical study in the Democratic Republic of the Congo to compare the performances, under field conditions, of the all-recombinant BIOLINE HAT 2.0, the native antigen SD BIOLINE HAT, and the traditional ‘standard of care’ CATT test [E3]. A total of 57,632 people were screened, and 260 HAT cases were confirmed by microscopy with benefit to the individuals involved who received treatment. The outcome was that the new BIOLINE HAT
2.0 device was more sensitive than SD BIOLINE HAT and CATT (71.2% versus 59.0% and 62.5%, respectively) while all three tests had a specificity above 98% [E3].
- Adoption and implementation of the new, supply chain-resilient HAT diagnostic
As a result of the definitive outcome of the clinical study, FIND officially adopted BIOLINE HAT
2.0 (Figure 1) as their preferred test for HAT screening and surveillance and launched the commercial product with Alere in September 2017 (Alere subsequently acquired by Abbott), acknowledging the contribution of the University of Dundee [E4]. The General Manager of SD Inc. said at the time:
“This test is the fruit of several years of joint efforts, and a major achievement resulting from the successful partnership that has been established with FIND to support the development of rapid tests for diseases associated with poverty” [E4].
The Chief Executive Officer of FIND said the test:
“will facilitate diagnosis of sleeping sickness patients in even the most remote corners of affected countries and enhance the prospects of achieving and sustaining elimination of the disease” [E4].
Figure 1. BIOLINE™ 2.0 Rapid diagnostic test (C = control; 2,1 = test antigens).
Full commercial roll out was anticipated to complete in spring 2020 but has been delayed due to FIND’s need to develop and market RDTs for COVID-19. The test is now planned to complete roll out in February 2021 [E2].
- Commissioning of a combined RDT prototype for both HAT and malaria
Patients with malaria or the early stage of HAT have similar symptoms. A diagnostic test that differentiates between them could inform treatment and control transmission. The success of BIOLINE HAT 2.0 led FIND to commission Abbott to develop a combined RDT incorporating Ferguson’s rISG65 recombinant antigen and an antigen from the malaria parasite. The new combined RDT is as accurate as the individual HAT and malaria RDTs under laboratory conditions [E5]. FIND has sponsored a new clinical observational study to evaluate the dual RTD in the field to screen for HAT and diagnose malaria [E6].
FIND has declared the transformational impact of the availability of BIOLINE HAT 2.0 and the development of the HAT/malaria combined RDT in their diagnostic strategy pipeline for HAT [E7]. The tests are FIND’s two prioritized activities for HAT in “ addressing the most critical gaps in the diagnostic landscape” for screening interventions (the other two tests are for case confirmation) [E7]. FIND’s HAT screening programme was supported by investment from the Bill & Melinda Gates Foundation, UK aid from the UK Government, and the Swiss Government [E4].
5. Sources to corroborate the impact
[E1] University of Dundee press release confirming signing of the Material Transfer Agreement with FIND whereby Dundee would provide their reagents to FIND for the development on an improved diagnostic for HAT, 22nd April 2014.
[E2] Corroboratory testimonial from Senior Project Manager of the Neglected Tropical Diseases Programme at FIND.
[E3] Clinical study reporting on evaluation of the diagnostic accuracy of BIOLINE HAT 2.0 (previously SD BIOLINE HAT 2.0) acknowledging role of Ferguson and citing publications R1 and R3. Lumbala, C, Biéler, S, Kayembe, S, Makabuza, J, Ongarello, S, Ndung’u, JM (2018) ‘Prospective evaluation of a rapid diagnostic test for Trypanosoma brucei gambiense infection developed using recombinant antigens’. PLoS Neglected Tropical Diseases, vol.12, no. 3, e0006386. DOI: 10.1371/journal.pntd.0006386
[E4] Press release from FIND announcing launch of second-generation rapid diagnostic test for sleeping sickness, 12th September 2017.
[E5] Report on the accuracy of the BIOLINE HAT/Malaria Combined prototype RDT. The Combo RDT uses Dundee rISG65. (SD BIOLINE HAT 2.0 used as a comparison here subsequently renamed BIOLINE HAT 2.0 by Abbott). Lumbala, C, Matovu, E, Sendagire, H, Kazibwe, AJN, Likwela, JL, Muhindo Mavoko, H, Kayembe, S, Lutumba, P, Biéler, S, Van Geertruyden, JP & Ndung'u JM. (2020) ‘Performance evaluation of a prototype rapid diagnostic test for combined detection of gambiense human African trypanosomiasis and malaria’. PLoS Neglected Tropical Diseases. 2020, vol.14, no.4, e0008168. DOI: 10.1371/journal.pntd.0008168
[E6] Clinical Study sponsored by FIND employing the BIOLINE HAT 2.0 (previously SD BIOLINE HAT 2.0) and BIOLINE HAT/Malaria Combo RTDs that use the Dundee rISG65. ClinicalTrials.gov (NCT003394976) 9th January 2018.
[E7] 2015-2020 ‘Strategy for Neglected Tropical Diseases’ FIND. pp 12-13.
- Submitting institution
- University of Dundee
- Unit of assessment
- 5 - Biological Sciences
- Summary impact type
- Technological
- Is this case study continued from a case study submitted in 2014?
- No
1. Summary of the impact
Visceral leishmaniasis is a risk to 600 million people with over 50,000 new cases annually, making it one of the world’s major parasitic killers. Research at the University of Dundee resulted in Phase II clinical trials to determine the efficacy of repurposing the drug fexinidazole to treat visceral leishmaniasis. Dundee drug discovery programmes, in collaboration with GlaxoSmithKline, have delivered two further novel preclinical drug candidates with different modes of action that are in clinical trials. The efficiency and productivity of leishmaniasis drug discovery in industry has been revolutionised by adoption of Dundee’s screening platforms and mode of action data.
2. Underpinning research
There are estimated to be 600 million people at risk of visceral leishmaniasis (VL) across the globe with 50,000 to 90,000 new cases every year, mainly among the poorest of the poor. The disease is caused by Leishmania parasites and is spread through the bite of infected sandflies. Limited therapeutic options make the treatment of this neglected disease very challenging. Each drug currently in use has serious drawbacks, such as difficulty in administration, length of treatment, toxicity, cost, and emerging drug resistance. For example, miltefosine is the only oral drug, but is contraindicated in women of childbearing age as it can cause birth defects.
In 2010, the global not-for-profit Drugs for Neglected Diseases initiative (DND i) reported ‘rediscovery’ of the abandoned drug fexinidazole for the treatment of Trypanosoma brucei (causing African sleeping sickness), a parasite related to Leishmania. The drug was thought to require activation by an enzyme in the parasite. Despite the prevailing consensus at DND i that fexinidazole was inactive against Leishmania, the Dundee team identified that Leishmania had a gene for a similar activating enzyme. In 2011, Prof Alan Fairlamb and Dr. Susan Wyllie in collaboration with Prof Kevin Read and the University of Dundee Drug Discovery Unit (DDU) undertook a project to examine the efficacy of fexinidazole against Leishmania donovani. This culminated in 2012 with the demonstration that fexinidazole displayed excellent parasite killing activity in vivo [R1] with activity dependent on oral administration and fexinidazole transformation in the liver into its active metabolites. The work was reported pre-publication to DND i and in 2013 they instigated a Phase II proof-of-concept clinical trial for the treatment of adults with VL in East Africa.
In parallel, given the paucity of front-line treatments for VL, the DDU in 2011 initiated a major research programme to uncover new drugs for this disease, including developing novel assay platforms to revolutionise leishmaniasis drug discovery [R2, R3]. A ‘Mode of Action’ research team was also established to discover the molecular targets of drug candidates that were at an advanced stage of development [R4-R6]. The DDU and GlaxoSmithKline currently have a research team of >40 scientists working on this Wellcome-funded programme of translational research for VL. Success of the programme is evidenced by the discovery and development of two clinical drug candidates for VL, each with different mechanisms of action. The first candidate arose from a compound series previously discovered in Dundee that inhibited growth of the related Trypanosoma brucei. Through clever iterative changes to the chemical core scaffold, the Dundee team were able to produce a compound active against the Leishmania parasite. The work, published in 2018, also identified the target of the compound as cyclin-dependent related kinase 12 (CRK12), providing Pharma with a completely novel validated drug target for Leishmania [R4]. Discovery of the second clinical drug candidate also began with compounds discovered in Dundee to be active against a different related parasite, Trypanosoma cruzi, which causes Chagas disease. Repurposing and optimisation in Dundee [R5, R6] produced a compound with potent activity against Leishmania that was shown to act through inhibition of the proteasome [R6].
3. References to the research
[R1] Wyllie, S, Patterson, S, Stojanovski, L, Simeons, FRC, Norval, S, Kime, R, Read, KD & Fairlamb, AH (2012) 'The Anti-Trypanosome Drug Fexinidazole Shows Potential for Treating Visceral Leishmaniasis', Science Translational Medicine, vol. 4, no. 119, pp119re1, DOI: 10.1126/scitranslmed.3003326
[R2] De Rycker, M, Hallyburton, I, Thomas, J, Campbell, L, Wyllie, S, Joshi, D, Cameron, S, Gilbert, IH, Wyatt, PG, Frearson, JA, Fairlamb, AH & Gray, DW (2013) 'Comparison of a high-throughput high-content intracellular Leishmania donovani assay with an axenic amastigote assay.', Antimicrobial Agents and Chemotherapy, vol. 57, no. 7, pp. 2913-2922. DOI: 10.1128/AAC.02398-12
[R3] Nühs, A, De Rycker, M, Manthri, S, Comer, E, Scherer, CA, Schreiber, SL, Ioset, J-R & Gray, DW (2015) 'Development and validation of a novel Leishmania donovani screening cascade for high-throughput screening using a novel axenic assay with high predictivity of Leishmanicidal intracellular activity', PLoS Neglected Tropical Diseases, vol. 9, no. 9, e0004094. DOI: 10.1371/journal.pntd.0004094
[R4] Wyllie, S, Thomas, M, Patterson, S, Crouch, S, De Rycker, M, Lowe, R, Gresham, S, Urbaniak, MD, Otto, TD, Stojanovski, L, Simeons, FRC, Manthri, S, MacLean, LM, Zuccotto, F, Homeyer, N, Pflaumer, H, Boesche, M, Sastry, L, Connolly, P, Albrecht, S, Berriman, M, Drewes, G, Gray, DW, Ghidelli-Disse, S, Dixon, S, Fiandor, JM, Wyatt, PG, Ferguson, MAJ, Fairlamb, AH, Miles, TJ, Read, KD & Gilbert, IH (2018) 'Cyclin-dependent kinase 12 is a drug target for visceral leishmaniasis', Nature, vol. 560, no. 7717, pp. 192-197. DOI: 10.1038/s41586-018-0356-z
[R5] Thomas, MG, De Rycker, M, Ajakane, M, Albrecht, S, Álvarez-Pedraglio, AI, Boesche, M, Brand, S, Campbell, L, Cantizani-Perez, J, Cleghorn, LAT, Copley, RCB, Crouch, SD, Daugan, A, Drewes, G, Ferrer, S, Ghidelli-Disse, S, Gonzalez, S, Gresham, SL, Hill, AP, Hindley, SJ, Lowe, RM, MacKenzie, CJ, MacLean, L, Manthri, S, Martin, F, Miguel-Siles, J, Nguyen, VL, Norval, S, Osuna-Cabello, M, Woodland, A, Patterson, S, Pena, I, Quesada-Campos, MT, Reid, IH, Revill, C, Riley, J, Ruiz-Gomez, JR, Shishikura, Y, Simeons, FRC, Smith, A, Smith, VC, Spinks, D, Stojanovski, L, Thomas, J, Thompson, S, Underwood, T, Gray, DW, Fiandor, JM, Gilbert, IH, Wyatt, PG, Read, KD & Miles, TJ (2019) 'Identification of GSK3186899/DDD853651 as a Preclinical Development Candidate for the Treatment of Visceral Leishmaniasis', Journal of Medicinal Chemistry, vol. 62, no. 3, pp. 1180-1202. DOI: 10.1021/acs.jmedchem.8b01218
[R6] Wyllie, S, Brand, S, Thomas, M, De Rycker, M, Chung, C-W, Peña, I, Bingham, R, Bueren-Calabuig, J, Cantizani, J, Cebrian, D, Craggs, PD, Ferguson, L, Goswami, P, Hobrath, J, Howe, J, Jeacock, L, Ko, EJ, Korczynska, J, MacLean, L, Manthri, S, Santos Martinez, M, Mata-Cantero, L, Moniz, S, Nuhs, A, Osuna-Cabello, M, Pinto, E, Riley, J, Robinson, S, Rowland, P, Simeons, F, Shishikura, Y, Spinks, D, Stojanovski, L, Thomas, J, Thompson, S, Viayna Gaza, E, Wall, R, Zuccotto, F, Horn, D, Ferguson, M, Fairlamb, A, Fiandor, JM, Martín, J, Gray, D, Miles, TJ, Gilbert, I, Read, K, Marco, M & Wyatt, PG (2019) 'Preclinical candidate for the treatment of visceral leishmaniasis that acts through proteasome inhibition', Proceedings of the National Academy of Sciences, vol. 116, no. 19, 201820175, pp. 9318-9323. DOI: 10.1073/pnas.1820175116
Key research grants relevant to this case study:
Fairlamb, AH. Characterization and validation of drug targets in the Kinetoplastida. Wellcome Principal Research Fellowship (2006-2016). Wellcome. Value: GBP 5,419,846.
Wyatt, PG. A pipeline of drugs for leishmaniasis and Chagas disease (2017-2022) Wellcome. Value: GBP 7,454,810.
Wyatt, PG Centre for Neglected Tropical Diseases Drug Discovery (2017-2022). Wellcome. Value: GBP 13,611,794.
Wyatt, PG Discovery and Development of Drug Candidates for Neglected Diseases. Strategic Award (2011-2017). Wellcome. Value: GBP 10,174,124.
Gilbert, IH Chemical biology: Leveraging phenotypic hits against kinetoplastids. (2015-2019). Wellcome. Value: GBP 2,301,742.
Gray DW Axenic assay development and screening of external compounds. Drugs for Neglected Diseases initiative (2012-2014). Value: GBP 179,000.
Wyatt, PG Identification of Drug Leads to Treat Leishmaniasis. Drugs for Neglected Diseases initiative (2009-2014). Value: GBP 1,800,000.
Gilbert, IH A Translational Engine for Biomedical Discoveries. Strategic Award (2013-2015). Wellcome. Value: GBP 800,000.
4. Details of the impact
Phase II clinical trial of fexinidazole for treatment of human VL, and application in dogs
The discovery by Fairlamb, Read and Wyllie that oral fexinidazole had anti-leishmania activity in mouse models prompted a re-evaluation by DND i that ultimately resulted in a Phase II clinical trial to determine the efficacy and safety of fexinidazole to treat VL patients [E1]. All patients benefited from clinical improvement during treatment and the trial delivered conclusive knowledge to DND i that fexinidazole reduces parasite burden. However, not all patients remained cured due to insufficient exposure to fexinidazole (requiring higher or longer dosing) and the trial terminated [E1]. Since anti-leishmanial drugs are subject to resistance, DND i’s goal is for two safe, orally administered drugs to be used in combination. Fexinidazole is one of only two orally-available therapies and it’s superior safety profile compared to the other oral drug, miltefosine, means it could be a partner candidate for combination treatments with new oral drugs in development.
Leishmania infantum is the causative agent of infantile VL in the Mediterranean region and Latin America, with expanding geographical incidence. Dogs are the primary reservoir for infection of humans and up to 10% of dogs in endemic areas develop disease. Existing treatments do not produce a reliable cure and have significant side effects. In 2014, Merial (now Boehringer Ingelheim Animal Health) filed a patent on fexinidazole as a treatment and cure for leishmaniasis in dogs, evidenced by trials they had undertaken [E2]. Their trials and patent are a clear direct consequence of the earlier published DDU research [R1] which they cite.
Delivery and clinical trial of new oral candidates for VL treatment in collaboration with GSK
There is an urgent need for new oral treatments for VL that work through novel modes of action. This enables combination therapies that will improve efficacy and reduce the risk of resistance. In April 2017, DND i and GlaxoSmithKline, in collaboration with the DDU, entered into a formal agreement for the preclinical development of two such compounds [R4, R5] discovered in Dundee for treatment of leishmaniasis [E3]. The most advanced compound was declared as a preclinical candidate by GlaxoSmithKline in July 2018 [E4] and human Phase 1 clinical trials began in May 2019 [E5]. The second compound was approved for Phase 1 clinical trials in June 2020 [E6] and the first subjects were dosed in October that year.
These clinical candidates represent step-changes in the development of treatments for VL since both are orally bioavailable in humans. Globally, only three other oral VL candidates are currently being clinically assessed [E7]. Dundee has therefore contributed 40% of the global new compounds in the pipeline for combination clinical testing. Addressing the impact of these new drug treatments, the Discovery Director at DND i said:
“To have a new compound with a novel mode of action against VL is a huge advance for the field as it enables testing and development of multiple new combination therapies with other new and existing drugs. For the DDU to identify not one, but two such new drug candidates in just a few years, is an outstanding achievement for an academic drug discovery unit” [E8].
Adoption of gold-standard screening assays for leishmaniasis drug development
An important impact of Dundee’s research has been adoption by industry of a suite of screening and assay platforms developed by the DDU for VL drug discovery. These were transferred to GlaxoSmithKline (following training of GSK staff in Dundee) and are now widely used by industry and Product Development Partnerships [E9]. In total, DNDi and GlaxoSmithKline have screened >500,000 and 1.8 million compounds respectively using DDU assays [E9], a feat previously impossible with earlier, laborious assays. DDU Mode of Action screening cascades also provided DND i, GlaxoSmithKline and others with knowledge that saved resources by enabling decisions not to proceed with compounds that might have negative outcomes [E10]. Overall, the DDU’s drug discovery expertise has enhanced the productivity of the global VL drug discovery portfolio. DND i comment on DDU’s gold standard screening assays thus:
“…continued development of high capacity, physiologically relevant assays using Leishmania by the Dundee DDU has been transformational for visceral leishmaniasis (VL) drug discovery. The high throughput [parasite-killing] assay and high content imaging platform for intracellular assays have revolutionized the capacity and efficiency of screening, enabling many millions of compounds to be screened for new chemical starting points…The Dundee L.donvani screening platform allowed rapid identification of hit series of molecules for both Dundee and other groups around the world, including major Pharma companies.” [E8].
5. Sources to corroborate the impact
[E1] (i) Powerpoint presentation from DNDi presenting data from R1.
(ii) Phase II Clinical Trial: (NCT01980199). 8th November 2013.
[E2] Patent for fexinidazole as treatment and cure of canine leishmaniasis citing Dundee underpinning research R1: PCT/US2014/014134 (WO2014121064A1).
[E3] DND i R&D Portfolio updates about the two new oral candidates for VL, GSK3186899/DDD853651 and GSK3494245/DDD1305143.
[E4] Press release citing R1 on declaration of preclinical drug candidate to treat VL, GSK3186899/DDD853651.
[E5] Phase I Clinical Trial (NCT03874234) 14th March 2019.
[E6] Phase I Clinical Trial (NCT04504435) 7th August 2020.
[E7] DND i 2020 portfolio update validating that Dundee’s GSK3186899/DDD853651 & GSK3494245/DDD1305143 are two of only five candidate drugs in Phase 1 for treatment of VL.
[E8] Corroboratory testimonial from Discovery Director, DND i.
[E9] DND i Visceral leishmaniasis R&D portfolio screening collaboration with Dundee to screen 500,000 compounds plus GlaxoSmithKline publication also using DDU assay to screen 1.8million compounds.
DND i R&D Portfolio 2020, ‘Screening leishmaniasis’ DNDi 29th Feb 2020.
Publication by GSK: Peña, I, Pilar Manzano, M, Cantizani, J, Kessler, A, Alonso-Padilla, J, Bardera, AI, Alvarez, E, Colmenarejo, G, Cotillo, I, Roquero, I, de Dios-Anton, F, Barroso, V, Rodriguez, A, Gray, DW, Navarro, M, Kumar, V, Sherstnev, A, Drewry, DH, Brown, JR, Fiandor, JM & Julio Martin, J 2015, 'New compound sets identified from high throughput phenotypic screening against three kinetoplastid parasites: an open resource', Scientific Reports, vol. 5, 8771. https://doi.org/10.1038/srep08771
[E10] Press release announcing the GlaxoSmithKline Scientific Termination of Projects (STOP) Award 2017 to the University of Dundee Mode of Action group for producing data that enabled de-prioritisation of inappropriate compound series aimed at VL.
- Submitting institution
- University of Dundee
- Unit of assessment
- 5 - Biological Sciences
- Summary impact type
- Technological
- Is this case study continued from a case study submitted in 2014?
- No
1. Summary of the impact
Exscientia, a spin-out company of the University of Dundee created by Prof Andrew Hopkins FRSE, has grown into a market leader in artificial intelligence drug design technology. Since 2014, the company has attracted GBP84 million investment, earned revenue of GBP23.9 million, created 88 highly-skilled jobs, and serviced 18 partnerships with major global Pharma and others (GBP538 million in disclosed deals, with more undisclosed). Exscientia has reduced the time and cost of pre-clinical drug discovery and has delivered two novel drugs into first-in-human clinical trials, being the fastest AI-design drug company to reach that critical stage.
2. Underpinning research
The world has rarely been more acutely aware than it is now of the time and cost involved in the development of new drugs and vaccines. The pharmaceutical Industry is facing serious pressures, given estimates of a USD2.6 billion cost and 10-15 year timeline for developing a new drug. Declining drug discovery R&D productivity is the most important challenge to address. One approach to tackle this problem is to harness chemical, pharmacological and biological data for automated drug design. Prof Andrew Hopkins has been at the forefront of this research by combining machine learning and big data analytics to tackle questions of target identification, polypharmacology and de novo compound design.
Hopkins is a pioneer of ‘network pharmacology’, coining the phrase in 2007 and expanding in a seminal 2008 review cited 1,324 times [R1]. Network pharmacology combines biological network analysis with chemical biology approaches, to design new drugs to perturb biological networks rather than individual targets. This is contrary to conventional drug discovery approaches based on highly specific targeting of a single protein that often lead to lower than desired clinical efficacy. Network pharmacology is the concept that drugs for many diseases may require multiple activities to be most effective. This research by Hopkins opened up a new subfield in drug discovery [R1].
The creation of ChEMBL in 2008, provided the first publicly-available database of bioactive molecules including pharmacological and chemical structural-activity relationship data that, significantly, was in a format amenable to reading by computers. Taking advantage of this resource, Hopkins embarked on a research project to harness the high-quality chemoinformatics data and apply machine-led learning to the process of new drug design. His team systematically integrated data derived from different disciplines including computational modelling [R2], synthetic chemistry, biophysics [R3], pharmacological testing and clinical studies, to develop methods and algorithms [R4, R5] to automate drug design. Hopkins collaborated with experimentalists at the University to efficiently design novel patentable chemicals for synthesis and then test the compounds designed by the algorithms [R2]. This resulted in creation of an automated, adaptive methodology for designing drug ligands to multi-target profiles, with a 75% prediction success rate verified experimentally and the predicted target engagement confirmed in vivo [R2-R6].
It was quickly realised that the computational technology platform developed by the University of Dundee offered a highly scalable system to generate intellectual property [R5,R6]. Spin-out Exscientia Ltd was created in 2012 as a technology platform company aiming to revolutionise productivity in drug discovery using data analytics and AI (Artificial Intelligence) techniques of machine learning during the initial phases from chemical design to production of a clinical candidate.
3. References to the research
[R1] Hopkins, AL (2008) 'Network pharmacology: the next paradigm in drug discovery', Nature Chemical Biology, vol. 4, no. 11, pp. 682-690. DOI: 10.1038/nchembio.118
This paper is the highest cited paper ever published in Nature Chemical Biology since 2008.
[R2] Besnard, J, Ruda, GF, Setola, V, Abecassis, K, Rodriguiz, RM, Huang, X-P, Norval, S, Sassano, MF, Shin, AI, Webster, LA, Simeons, FRC, Stojanovski, L, Prat, A, Seidah, NG, Constam, DB, Bickerton, GR, Read, KD, Wetsel, WC, Gilbert, IH, Roth, BL & Hopkins, AL (2012) 'Automated design of ligands to polypharmacological profiles', Nature, vol. 492, no. 7428, pp. 215-220. DOI: 10.1038/nature11691 This paper received significant coverage, with News and Views features in Nature Reviews Drug Discovery and C&E News and in the press (including the BBC https://bbc.in/3e5K0Ks)
[R3] Navratilova, I & Hopkins, AL (2010) 'Fragment Screening by Surface Plasmon Resonance', ACS Medicinal Chemistry Letters, vol. 1, no. 1, pp. 44-48. DOI: 10.1021/ml900002k
[R4] Bickerton, GR, Paolini, GV, Besnard, J, Muresan, S & Hopkins, AL (2012) 'Quantifying the chemical beauty of drugs', Nature Chemistry, vol. 4, no. 2, pp. 90-98. DOI: 10.1038/NCHEM.1243
[R5] Patent: Besnard, Jeremy and Hopkins, Andrew Lee. (2011) Design of Molecules. World Intellectual Property Organization Patent no. PCT/GB2010/051940 (WO2011061548). Available at https://bit.ly/2Oi2YTK
[R6] Patent: Besnard, Jeremy, Hopkins Andrew Lee, Gilbert, Ian, Ruda Gian Filippo, Abecassis, Keren (2012) *Morpholino compounds, uses and methods **. World Intellectual Property Organization Patent no. PCT/GB2012/051194 (WO2012160392) Available at https://bit.ly/3qu0EGp
Key research grants relevant to this case study:
Hopkins, AL Exscientia: Commercialising Multi-Target Drug Design. BBSRC Follow on Fund (2011- 2012). Value: GBP150,100
Hopkins, AL Experimental Proof of Concept for Drug Design Selective Optimisation Algorithm. BBSRC Pathfinder Award for Drug Design Proof-of-Concept (2009-2010). Value GBP13,759
4. Details of the impact
a) Raising multi-million investment in, and revenue from, AI-guided drug design
Exscientia has rapidly-grown into a leading pharmatech company at the forefront of AI drug discovery. Since 2017, Exscientia has raised a total of GBP84 million, comprising:
Series A EUR15 million in 2017 [E1]
Series B USD26 million in 2019 [E2]
Series C USD70 million in 2020 [E3]
(including USD10 million raised after the announcement).
Evotec, one of four investors, said of Series A in 2017:
“Our investment in Exscientia represents Evotec’s single biggest equity placement to date and in, what we feel, is the world leading AI technology company” [E1].
Exscientia’s revenue rose from GBP1.5 million in 2018 to GBP9.8 million in 2020, with GBP23.0 million total revenue since 2016 [E4]. This enabled Exscientia to expand from its initial space in Dundee, to a second office in Oxford (2017), then into larger premises on the Oxford Science Park (2018). Recently (2019/2020), offices opened in Osaka (Japan) and Miami (USA). In the REF period, expansion increased the workforce from 3 to 91 staff [E4] by creating 88 highly-skilled new jobs (headcount: 88; FTEs: 88) with staff numbers doubling each year for the past 3 years (Figure).
b) Innovation and partnership with Pharma to accelerate drug discovery using AI
In addition to its own pipeline, Exscientia provides services to pharmaceutical companies where its automated AI drug design process can accelerate development of candidates for clinical trial.
These services have attracted 18 partnerships on 28 separate projects, developing drugs or technology for a range of diseases, including cancer, COVID-19, psychiatric disease, bone disease, fibrosis and others [E4]. Many collaborations are with global Pharma companies with revenue generated through initial upfront and research payments, near-term and clinical milestone payments, and royalties on final products. Examples of such deals include: Bayer (2019; EUR240 million), Roche (2019; CHF67 million), Celgene/Bristol Myers Squibb (2019; USD25 million upfront, with undisclosed milestones), GlaxoSmithKline (2017; GBP33 million), Sanofi (2017, EUR250 million), Sunovion (2014; total USD4.8 million) [E4]. Partnerships also exist with GT Apeiron, Evotec, SRI, RallyBio, Blue Oak, Huadong Medicine, IMI CARE, Diamond Light Source/Calibr, Sumitomo Dainippon Pharma, and the Gates Foundation [E4]. Twelve patents were filed or published on Exscientia molecules/methods in the REF period [E4].
c) Significant efficiency and cost savings compared to conventional drug discovery
Published industry benchmarks estimate it takes 4.5 years and USD63 million to get to pre-clinical testing of a drug candidate. In 2014, Exscientia signed a partnership with Sumitomo Dainippon Pharma Ltd and one year later delivered a molecule meeting Sumitomo criteria [E5]. Fewer than 400 compounds needed to be synthesized to identify the final compound using Exscientia’s automated system, saving over 80% costs in these drug discovery stages, or 30% of the total drug development cost [E4, E6]. The Director of Sumitomo’s chemistry team said:
“Working with Exscientia has been transformative, enabling us to progress projects far faster than had previously been possible”, and their Director of Drug Development added, “... projects by ex scientia are providing exciting productivity and efficiency gains…” [E5].
In 2019, Exscientia provided GlaxoSmithKline with a first selective, potent in vivo, lead molecule, discovered in 5 iterative cycles with only 85 compounds tested [E7] in comparison with industry benchmarks of 2,500 per project. Also in 2019, Sanofi exercised its option to advance a compound from their 2017 partnership where Exscientia designed bispecific-binding molecules after triaging >1,000 disease-relevant target combinations and designing nearly 100 billion novel compounds. The first-in-class bispecific molecule being progressed by Sanofi interacts with two drug targets related to inflammation and fibrosis - normally needing two separate drugs [E8].
These achievements highlight the efficiency and productivity gains made possible through Exscientia’s algorithms. Their current pipeline has 12 drug discovery projects [E4], and 5 new drug candidates have been delivered in under 14 months, far quicker than the 4.5 to 5-year industry benchmark and with associated cost savings [E7]. Evotec confirm:
“Through our partnership with Exscientia we have seen first-hand evidence that they can deliver the most productive drug discovery engine in the industry” [E2].
d) Fastest AI-designed drugs to accelerated IP filing and first-in-human clinical trials
Exscientia has been the fastest AI-design drug developer in the world to reach the clinic, enabling accelerated IP filing. In January 2020, though partnership with Sumitomo Dainippon Pharma Ltd, Exscientia delivered into Phase 1 clinical trials the first AI-designed drug ever to be tested in humans [E9]. The drug, in trial in Japan, treats patients with obsessive-compulsive disorder, enabling Sumitomo to expand its pipeline to treat key unmet needs in psychiatry/neurology. Sumitomo’s Senior Executive Officer said:
“We are very excited with the results of the joint research that resulted in the development of candidate compounds in a very short time…” [E9].
An Exscientia-proprietary AI-designed drug targeting cancer is also in Phase I first-in-human clinical trial, enrolling its first participant in December 2020 [E9]. Exscientia and Hopkins have received numerous accolades (collated in E10) and their leading position in the sector is confirmed by many:
GSK say “ Exscientia has… proven innovation in drug discovery technologies…their industry-leading approach will accelerate the discovery of new molecules” [E4];
Forbes list Exscientia as a “ leading AI drug discovery startup” for possible acquisition by Big Pharma for over USD2 billion in the near future [E10];
Evotec say “[Exscientia’s] AI approaches…can positively and radically impact drug discovery” [E1].
5. Sources to corroborate the impact
[E1] Press Release on Investment by Evotec. Financial Times, ‘Exscientia announces investment from Evotec’. Frontier IP Group PLC. 28th September 2017.
[E2] Press Release on Series B funding round and Roche partnership. Financial Times, ‘Exscientia Series B funding & Roche collaboration’ Frontier IP Group PLC. 7th January 2019.
[E3] Series C funding announcement, ‘Novo Holdings leads USD60m Series C financing round in Exscientia’. Novo Holdings. 26th May 2020.
[E4] Corroboratory testimonial statement from Exscientia and collated partnership deal press releases.
[E5] Exscientia statement on partnership with Sumitomo Dainippon Pharma 2015, ‘Exscientia Ltd, reaches first delivery milestone in collaboration with Sumitomo Dainippon Pharma Co., Ltd’. Exscientia 2nd September 2015. Available at https://bit.ly/37vvc4l
[E6] Exscientia featured in Deloitte Insights report. Deloitte Insights 2019, ‘Intelligent drug discovery’ Deloitte.com pp21-23. 7th November 2019 Available at https://bit.ly/3mxplj0
[E7] Press release on delivery of preclinical candidate for GSK. Financial Times, ‘Exscientia reaches first major milestone with GSK’. Frontier IP Group PLC. 4th April 2019
[E8] Press release on product development and license option agreement with Sanofi. Exscientia, ‘Sanofi Exercises option on Exscientia-designed bispecific small molecule for treatment of immunological conditions’ Exscientia.ai 5th August 2019.
[E9] (i) Financial Times article on first AI designed drug to enter human clinical trials. Murgia M 2020 ‘AI-designed drug to enter human clinical trial for first time’. Financial Times. 30th January 2020. Available at https://on.ft.com/2KfBDzp; and Press Release 2020, ‘Exscientia: world first trials of new drug candidate created by artificial intelligence’ Frontier IP Group plc. 31st January 2020. Available at https://bit.ly/3paWaDE
(ii) Phase 1 Clinical Trial of Exscientia drug EXS21546. Sponsor: Exscientia 2020, ‘3-part Study to Assess Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of EXS21546’ ClinicalTrials.gov (NCT04727138). 8th December 2020. Available at https://bit.ly/3a4LTol
[E10] Compilation of external accolades and awards.
2015 BBSRC Commercial Innovator of the Year
2015 Scottish Enterprise award for Life Sciences Entrepreneurial Business Leadership
2017 OBN Awards Best Emerging Biotech Company
2017 RSC Chemistry World Entrepreneur of the Year
2019 Lifestar Awards Series B Finance Raise of the Year
2019 Top 20 most promising world companies using AI drug discovery (American Chemical Society Discovery Report page 13)
2020 Sunday Times Tech Track 100 league table
2020 OBN Awards Best Established Biotech company finalist
Condie B & Dayton L 2020, ‘Four AI technologies that could change the way we live and work’ Nature.com 9th December 2020. Available at https://bit.ly/37r6dyS
Toews R 2020, ‘10 AI Predictions for 2021’ Forbes 22 December 2020. Available at https://www.forbes.com/sites/robtoews/2020/12/22/10-ai-predictions-for-2021/?sh=4c45ab05d1a0
- Submitting institution
- University of Dundee
- Unit of assessment
- 5 - Biological Sciences
- Summary impact type
- Health
- Is this case study continued from a case study submitted in 2014?
- No
1. Summary of the impact
Malaria kills 400,000 people annually, the majority being children. Resistance to current treatments has generated an urgent need for new drugs effective across different parasite life-cycle stages, to enable malaria treatment and eradication. Research at the University of Dundee’s Drug Discovery Unit has resulted in the invention of a novel small molecule with demonstrated ability to cure malaria, prevent infection, and block transmission with a single dose. This research:
de-risked pre-clinical drug discovery, incentivising partnership between Medicines for Malaria Venture and Merck for clinical development of the Dundee molecule;
led to a successful first-in-human clinical trial with accelerated volunteer infection study;
uncovered a novel mode of action, unblocking the pipeline for crucial combination treatments to combat malaria.
2. Underpinning research
Malaria is a debilitating parasitic disease posing a risk to nearly half the world’s population. The WHO estimated 229 million cases of malaria in 2018, with 409,000 deaths, most in young children and pregnant women in sub-Saharan Africa. Disruption of malaria treatment services due to COVID-19 is likely to increase malaria deaths in 2020. The predominant malaria parasite, Plasmodium falciparum, has developed resistance to many drugs, including the core artemisinin component of current first-line therapies. New drugs are urgently needed to effectively treat and eventually eradicate malaria. New anti-malarials must meet several requirements: (a) novel modes of action with no cross-resistance to current drugs; (b) single-dose treatments with activity against blood-stage disease; (c) activity against liver stages that can prevent disease development (chemoprotection or prophylaxis); (d) compounds active against the sexual stages (gametocytes) to prevent transmission of malaria. DDD107498, invented by researchers in the Drug Discovery Unit (DDU) at the University of Dundee, has a completely novel mode of action and meets all these criteria. It is one of only three compounds with this profile undergoing clinical development by the Medicines for Malaria Venture (MMV) not-for-profit public-private R&D partnership.
Invention of DDD107498 began by screening a Dundee in-house chemical compound library [R1] to identify compounds that inhibited growth of P. falciparum in human red blood cells. The screening tested 4,731 compounds and succeeded in identifying a promising compound series [R2]. A subsequent DDU medicinal chemistry programme optimized properties of the compound series through iterative cycles of designing improvements, making the improved compounds, and biological testing of their potency and metabolic stability [R3]. Further rounds of the design-make-test cycle improved their oral bioavailability, permeability, potency and selectivity. This remarkable chemical evolution transformed a series with suboptimal properties into a pre-clinical candidate with approximately 100-fold increase in potency and much improved chemical properties [R4]. The final compound was active against drug-resistant parasites with no cross-resistance to current antimalarial drugs. It had remarkable potency, oral bioavailability and a long half-life in malaria mouse models across multiple life cycle stages; cured blood stream infection; had crucial transmission-blocking activity ; and could even prevent infection in the first place [R3, R4].
The DDU, together with collaborators, identified that the compound acted through inhibition of protein synthesis, with parasite translation elongation factor eEF2 as its target [R4]. After treatment with DDD107498 and consequent inhibition of eEF2, the parasites cannot make essential proteins, and die. This discovery confirmed DDD107498’s completely novel mode of action, one that parasites will not have had a chance to develop resistance to, making it a very attractive prospect for further development. This work, published in Nature in 2015, is highly cited and received significant media coverage [R4]. A patent was filed around the compound series [R5].
This huge success in going from fundamental discovery science to a drug in successful clinical trial, highlights the unique on-going capability and impact of the DDU in translating world-class discovery research into new de-risked targets and candidate drugs. Its continuing malaria drug discovery effort has now identified a second compound series with a novel mode of action that has provided additional molecules to the malaria drug discovery pipeline.
3. References to the research
[R1] Brenk, R, Schipani, A, James, D, Krasowski, A, Gilbert, IH, Frearson, J & Wyatt, PG (2008) 'Lessons learnt from assembling screening libraries for drug discovery for neglected diseases', ChemMedChem., vol. 3, no. 3, pp. 435-444. DOI: 10.1002/cmdc.200700139
[R2] Hallyburton, I, Grimaldi, R, Woodland, A, Baragana, B, Luksch, T, Spinks, D, James, D, Leroy, D, Waterson, D, Fairlamb, AH, Wyatt, PG, Gilbert, IH & Frearson, JA (2017) 'Screening a protein kinase inhibitor library against Plasmodium falciparum', Malaria Journal, vol. 16, 446, pp. 1-11. DOI: 10.1186/s12936-017-2085-4
[R3] Baragaña, B, Norcross, NR, Wilson, C, Porzelle, A, Hallyburton, I, Grimaldi, R, Osuna-Cabello, M, Norval, S, Riley, J, Stojanovski, L, Simeons, FRC, Wyatt, PG, Delves, MJ, Meister, S, Duffy, S, Avery, VM, Winzeler, EA, Sinden, RE, Wittlin, S, Frearson, JA, Gray, DW, Fairlamb, AH, Waterson, D, Campbell, SF, Willis, P, Read, KD & Gilbert, IH (2016) 'Discovery of a quinoline-4-carboxamide derivative with a novel mechanism of action, multistage antimalarial activity, and potent in vivo efficacy' Journal of Medicinal Chemistry, vol. 59, no. 21, pp. 9672-9685. DOI: 10.1021/acs.jmedchem.6b00723
[R4] Baragaña, B, Hallyburton, I, Lee, MCS, Norcross, NR, Grimaldi, R, Otto, TD, Proto, WR, Blagborough, AM, Meister, S, Wirjanata, G, Ruecker, A, Upton, LM, Abraham, TS, Almeida, MJ, Pradhan, A, Porzelle, A, Santos Martinez, M, Bolscher, JM, Woodland, A, Luksch, T, Norval, S, Zuccotto, F, Thomas, J, Simeons, F, Stojanovski, L, Osuna-Cabello, M, Brock, PM, Churcher, TS, Sala, KA, Zakutansky, SE, Belén Jiménez-Díaz, M, Maria Sanz, L, Riley, J, Basak, R, Campbell, M, Avery, VM, Sauerwein, RW, Dechering, KJ, Noviyanti, R, Campo, B, Frearson, JA, Angulo-Barturen, I, Ferrer-Bazaga, S, Javier Gamo, F, Wyatt, PG, Leroy, D, Siegl, P, Delves, MJ, Kyle, DE, Wittlin, S, Marfurt, J, Price, RN, Sinden, RE, Winzeler, EA, Charman, SA, Bebrevska, L, Gray, DW, Campbell, S, Fairlamb, AH, Willis, PA, Rayner, JC, Fidock, DA, Read, KD & **Gilbert, IH (**2015) 'A novel multiple-stage antimalarial agent that inhibits protein synthesis' Nature, vol. 522, no. 7556, pp. 315-320. DOI: 10.1038/nature14451
[R5] Patent: Gilbert, Ian Hugh, Norcross, Neil, Baragaña, Beatriz, Porzelle, Achim (2013). Anti-malarial agents World Intellectual Property Organization. Patent No. PCT/GB2013/050633 (WO2013153357). Available at: https://bit.ly/2ONlQdm (Accessed:27 Nov 2020)
Key research grants relevant to this case study:
Frearson, J.A. Hit discovery in Plasmodium falciparum. Medicines for Malaria Venture (2008-2009). Value GBP40,112
Gilbert, I.H. Discovery and Optimization of Phenotypic Hits. Medicines for Malaria Venture (2010-2013). Value GBP1,192,000
Gilbert, I.H. Development of a Screen for the Identification of Plasmodium Falciparum eEF2 Inhibitors. Medicines for Malaria Venture (2016-2017). GBP99,820
4. Details of the impact
Single dose cure molecule targeting multiple lifecycle stages adopted by MMV
DDU research led to invention of DDD107498 that can treat malaria with a single dose. Crucially, it is effective against parasites resistant to current drugs at a dose within MMV’s goal of ~1USD per treatment, affordable in lower-income countries. In 2014, MMV formally declared DDD107498 as a candidate for preclinical development [E1]. The discovery won MMV’s Project of the Year 2014 [E2]. At the time MMV said:
“This molecule has caused a stir…. DDD498 has potent activity against multiple stages of the malaria parasite’s lifecycle, giving it the potential to cure and stop the spread of the disease as well as protect people, all in a single-exposure” [E2].
Incentivising Pharma by de-risking drug discovery
Developing a new drug costs over GBP1billion. For diseases like malaria, with high prevalence in developing countries, this investment is too high for Pharma, since it cannot be recouped from low-income patients or aid budgets. The DDU’s research to invent and characterize DDD107498 incentivized MMV to take it forward through a formal licensing partnership [E4]. In 2015 MMV explained the impact on them:
“Thanks to these attractive properties, MMV was able to successfully partner this compound with Merck KGaA…it was licensed their flagship anti-malarial for pre-clinical and clinical development and renamed M5717” [E3].
Similarly, the Head of Merck Global Health Institute explains the impact on the company:
“The DDU’s work to identify DDD107498 and characterise it and its safety profile meant that Merck were provided with a highly effective drug candidate ready for clinical trial…The DDU de-risked the process and provided the incentive for us to proceed to clinical development in 2017 with a new drug for malaria that we would have been very unlikely to develop independently. This also allowed Merck to develop our malaria portfolio” [E5].
Both MMV and Merck benefited by progressing their objective of defeating malaria, thereby encouraging sustained investment. People in malaria-endemic countries benefit from survival of such R&D partnerships working toward malaria treatments and eradication.
Malaria complete cure in clinical trial with first ever Phase 1 Volunteer Infection Study
Merck wished to fully understand M5717 (formerly called DDD107498) efficacy earlier than previously possible, due to its promise, and to minimise costs. In 2017, they began novel first-in-human Phase I trials that included volunteer infection studies (VIS) where healthy volunteers received a small inoculum of malaria to investigate M5717 as a clinical cure [E6]. This was the first time VIS were conducted by a pharmaceutical company as part of Phase I trials. The Head of Merck Global Health Institute confirmed the benefits of this unique study design, prompted by M5717’s promise/properties, saying that it “allowed us to reduce timelines, manage resources and minimize the number of patients needed in Phase II” [E5].
Merck reported M5717 as a single dose cure for malaria at the ASTMH Annual Meeting in 2019, following completion of the Phase I VIS trial. Their abstract confirms that “Administration of 800 mg M5717 resulted in complete clearance of parasitemia” [E7, abstract 1] . In a recent testimonial [E5] Merck confirm the trial demonstrated that M5717:
“…could cure subjects with a single dose, which marks a key milestone in its development…The phase I study demonstrated that M5717 has a positive benefit/risk profile and supports its further clinical development as a single dose for the treatment of malaria. M5717 is also active against the liver-stage of the parasite, making it a unique candidate for the prophylaxis and prevention of malaria.”
To this end, a second human challenge Phase 1b trial, initially delayed by COVID-19, has begun to assess the potential not just for cure but for prevention of malaria [E8]. The first subject was dosed in September 2020 [E9] (trial completes May 2021).
Unblocking the pipeline of new malaria combination treatments
WHO recommends combination antimalarial therapy, where each drug has a different mode of action to mitigate against the emergence of resistance. M5717’s completely novel mode of action opens up a pipeline towards new malaria combination treatments that partner it with existing or new drugs, and also identifies protein translation/eEF2 as targets for new drug development. Consequent impacts are the sale of DDD107498 to global customers by several international companies to facilitate drug discovery research [E10], and pursuit of combination clinical trials as confirmed by the Head of Merck Global Health Institute:
”*M5717 is identified as a key asset of the global malaria portfolio and is a central element of the next generation of malaria combinations that are managed by MMV. We are now in the process of identifying a potential partner drug to take M5717 further into Phase IIa.*” [E5].
The Chief Scientific Officer of MMV also explains:
“The fact that no-one had previously proposed EF2 as a target for malaria underlines the ground-breaking nature of this discovery…The compounds inhibiting EF2…are the only series we have seen so far…to hit all three [parasite] stages equally. This means that any drug will have potential not only for treating malaria, but for protecting against infection, and also to impact transmission… MMV are pleased to acknowledge the significant contribution and impact of the University of Dundee’s Drug Discovery Unit in delivering next-generation anti-malarial therapies. The combination of scientific rigour, the entrepreneurial spirit and the deep expertise… has made DDU a triumph of UK science over the last decade…” [E3].
5. Sources to corroborate the impact
[E1] Press release: Medicines for Malaria Venture 2014, ‘Potential new antimalarial drug identified at the University of Dundee’ MMV.org. 6th February 2014.
[E2] Press release: Medicines for Malaria Venture 2015, ‘MMV Project of the Year award 2014 - DDD498’ MMV.org. 30th June 2015.
[E3] Corroboratory testimonial from the Chief Scientific Officer, Medicines for Malaria Venture.
[E4] Press release: Medicines for Malaria Venture 2015, ‘Merck Serono and MMV sign agreement to develop potential antimalarial therapy’ MMV.org. 1st April 2015.
[E5] Corroboratory testimonial from the Head of Merck Global Health Institute, Merck KGaA and similar interview from 2018. Medicines for Malaria Venture 2018 ‘M5717 (formerly DDD498)’ MMV.org.
[E6] Clinical Trial of M5717: Sponsor Merck KGaA 2017, ‘First-in-Human Trial of Single Ascending Dose, Multiple Ascending Dose and Malaria Challenge Model in Healthy Subjects’ 25th Clinical Trials.gov (NCT03261401) 25th August 2017.
[E7] Corroboration of Phase 1b VIS clinical trial result: McCarthy J, Bagchus, W, Odedra, A, Webster, R, Oeuvray, C, Tappert, A, Bezuidenhout, D, Yin, X, Khandelwal, A & Yalkinoglu, O. 2019 ‘A Phase 1b study to investigate the antimalarial activity of m5717, a first-in-class inhibitor of plasmodium elongation factor 2, using the induced blood stage plasmodium falciparum malaria model’ ASTMH 68th Annual Meeting Abstract 1 p 1.
[E8] Clinical Trial: Sponsor Merck KGaA 2020, ‘Chemoprophylactic Activity of M5717 in Plasmodium Falciparum Sporozoite (PfSPZ) Challenge Model’ ClinicalTrials.gov (NCT04250363) 31st January 2020.
[E9] News Item: Merck Global Health Institute 2020 ‘Path to Zero Malaria: M5717 for prevention and cure of this deadly disease’ MerckGlobalHealthInstitute.com 8th September 2020.
[E10] Global Life sciences companies offering DDD107498 for commercial sale and citing [R4]: TargetMol Corp. DDD107498, Cat No T5419; ApexBio Technology LLC. DDD107498 Cat No A8711; MedChemExpress. DDD107498, Cat No. HY-117684A ; MedKoo Bioscience Inc. DDD107498, Cat No. 526858. Medkoo.com.
- Submitting institution
- University of Dundee
- Unit of assessment
- 5 - Biological Sciences
- Summary impact type
- Technological
- Is this case study continued from a case study submitted in 2014?
- No
1. Summary of the impact
Professor Alessio Ciulli FRSC is one of the pioneers of the development of a new class of drugs, the proteolysis-targeting chimeras (PROTACs), that target disease-causing proteins for degradation. Amphista Therapeutics, a University of Dundee 2017 spin-out, is based on his technology. PROTACs have changed the direction of commercial drug discovery programmes. Most global pharmaceutical companies today work on this revolutionary approach, prompting over USD3.5 billion investment in the targeted protein degradation sector. Ciulli’s structure-based design accelerates PROTAC deployment against previously undruggable disease proteins, as validated in a multi-million partnership with Boehringer Ingelheim. Benchmark PROTACs developed by Ciulli are marketed by several companies or freely-provided by Boehringer to speed innovation.
2. Underpinning research
Research led by Ciulli focussing on targeted protein degradation has driven the development of proteolysis-targeting chimeras (PROTACs). PROTACs are designed to harness the cell’s natural disposal system to specifically remove disease-causing proteins. A PROTAC is a two-headed molecule, where one end binds an enzyme (an E3 ligase) and the other binds the disease protein, bringing the two into close proximity. The ligase can then label the disease protein for degradation by the cell’s disposal system. Whereas conventional drugs only temporarily inhibit disease proteins by binding to their most important functional parts, PROTACs can bind at many positions and ensure the protein’s destruction. This revolutionary mode of action allows PROTACs to attack targets previously thought ‘undruggable’ and constitutes a platform technology applicable across diverse diseases.
The broad idea behind PROTACs was first suggested in 2001 by Crews (Yale) and Deshaies (California Institute of Technology) but early molecules did not attract drug companies as they relied on bulky peptides to bind the ligase, had low potency and could not easily enter cells. Collaboration between Ciulli (then at University of Cambridge) and Crews between 2010 and 2012 produced three publications identifying non-peptide molecules that bind the E3 ligase enzyme VHL (von Hippel-Lindau protein), albeit with only moderate potency, a limitation also in contemporary work with other ligases. In April 2013, Ciulli moved to the University of Dundee and in 2014 he published the structure-guided design and optimization of dramatically improved drug-like small molecule ligands that bind VHL with high potency [R1]. These were subsequently refined into a ligand with high VHL-specificity and cellular activity [R2].
In 2015, Ciulli developed one of the first ever non-peptidic two-headed PROTAC approaches by connecting his VHL ligands to a molecule that targets and binds to the BET (Bromodomain and Extra-Terminal motif) proteins that are critical for the growth and survival of cancer cells. This breakthrough cell-penetrant PROTAC, called MZ1, showed potent and unexpectedly selective degradation of one BET protein (called BRD4) over other BETs in cancer cells, offering unprecedented advantages over non-selective conventional inhibitors [R3]. As the BETs play crucial roles in pathways relevant to health and disease, the potential to accelerate development of drugs with minimal side effects and toxicity was immediately recognised.
Ciulli suggested that the specificity of a PROTAC might be influenced by the structure and interactions within each 3-part complex of ligase-PROTAC-target and, in 2017, he solved the first crystal structure of such a complex [R4]. This major breakthrough gave the new field the first ever glimpse of how a PROTAC brings the E3 ligase and its target protein together. Using this knowledge and the crystal structure to guide design, the Ciulli group produced a new PROTAC that had improved selectivity at depleting BRD4 [R4].
E3 ligases can themselves be disease-related proteins. Ciulli recently reported the first ‘Homo-PROTAC’ that can dimerize E3 ligase VHL, inducing self-destruction - a highly innovative strategy of ‘degrading the degrader’ [R5]. He has also designed novel small molecule VHL ligands [R6].
3. References to the research
[R1] Galdeano, C, Gadd, MS, Soares, P , Scaffidi, S, Van Molle, I, Birced, I, Hewitt, S, Dias, DM & **Ciulli, A (**2014) 'Structure-guided design and optimization of small molecules targeting the protein-protein interaction between the von Hippel-Lindau (VHL) E3 ubiquitin ligase and the hypoxia inducible factor (HIF) alpha subunit with in vitro nanomolar affinities' Journal of Medicinal Chemistry, vol. 57, no. 20, pp. 8657-8663. DOI: 10.1021/jm5011258
[R2] Frost, J, Galdeano, C, Soares, P, Gadd, MS, Grzes, KM, Ellis, L, Epemolu, O, Shimamura, S, Bantscheff, M, Grandi, P, Read, KD, Cantrell, DA, Rocha, S & **Ciulli, A (**2016) 'Potent and selective chemical probe of hypoxic signaling downstream of HIF-α hydroxylation via VHL inhibition', Nature Communications, vol. 7, 13312, pp. 1-12. DOI: 10.1038/ncomms13312
[R3] Zengerle, M, Chan, K-H & **Ciulli, A (**2015) 'Selective small molecule induced degradation of the BET bromodomain protein BRD4', ACS Chemical Biology, vol. 10, no. 8, pp. 1770-1777. DOI: 10.1021/acschembio.5b00216
[R4] Gadd, M, Testa, A, Lucas, X, Chan, KH, Chen, W, Lamont, D, Zengerle, M & Ciulli, A (2017) 'Structural basis of PROTAC cooperative recognition for selective protein degradation', Nature Chemical Biology, vol. 13, no. 5, pp. 514-521. DOI: 10.1038/nchembio.2329
[R5] Maniaci, C, Hughes, SJ, Testa, A, Chen, W, Lamont, DJ, Rocha, S, Alessi, DR, Romeo, R & **Ciulli, A (**2017) 'Homo-PROTACs: bivalent small-molecule dimerizers of the VHL E3 ubiquitin ligase to induce self-degradation', Nature Communications, vol. 8, 830, pp. 1-14. DOI: 10.1038/s41467-017-00954-1
[R6] Testa, A, Lucas, X, Castro, G , Chan, KH, Wright, J, Runcie, A , Gadd, M, Harrison, WTA, Ko, EJ, Fletcher, D & **Ciulli, A (**2018) '3-Fluoro-4-hydroxyprolines: Synthesis, conformational analysis and stereoselective recognition by the VHL E3 ubiquitin ligase for targeted protein degradation', Journal of the American Chemical Society, vol. 140, no. 29, pp. 9299-9313. DOI: 10.1021/jacs.8b05807
Key research grants relevant to this case study:
Ciulli, A. A Systems Approach for the Fragment-Based Development of Selective Chemical Probes of Bromodomain Function’, BBSRC (2013-2015). Award Value: GBP333,693
Ciulli, A. Dissecting and Exploiting Molecular Recognition at Protein-Protein Interfaces’. BBSRC (2013-2015). Award Value: GBP307,126
Ciulli, A. Probing Druggability of Multisubunit Complexes: E3 Cullin RING Ligases’ European Research Council (2013 –2018). Award value: EUR1,499,904
4. Details of the impact
Reshaping drug discovery programmes and investment within Pharma
Ciulli’s research helped transform a niche area of chemical biology into one of the most exciting new fields of drug discovery. An expert opinion piece in Nature [E1] cites his 2015 publication [R3] and work of two other groups as achieving the spectacular leap in performance that enabled PROTAC technology, which had not gained traction in a decade, to become the focus of a “ gold rush”. By 2019, PROTACs were “ driving billions of US dollars in investment from pharmaceutical companies such as Roche, Pfizer, Merck, Novartis and GlaxoSmithKline” [E1]. Intellectual property underpins investments; Ciulli has filed 3 patents since 2015 and 34 other unique patent families from 15 companies cite his research (including Arvinas, C4 Therapeutics, AstaZeneca, GlaxoSmithKline and Boehringer Ingelheim) [E2]. A 2020 global market report by Roots Analysis indicates over USD3.5billion investment in targeted protein degrader drugs since 2014, with PROTACs accounting for over 30% of pipeline drugs in this class.
Enabling development and successful clinical trial of PROTAC drugs for cancer patients
These research advances and investments directly resulted in a first wave of PROTAC drugs entering clinical trials. Arvinas Inc. has PROTACs in expedited Phase 1/2 clinical trials for highly refractory metastatic castration-resistant prostate cancer (>250,000 new patients a year in the USA alone) and metastatic breast cancer (half a million global deaths annually) respectively. In their December 2020 Clinical Program Update, Arvinas report continued patient benefit and clear signs of efficacy. The development of these drugs was significantly dependent on the research contribution of Ciulli, with R1-R6 variously cited in 11 patent families filed by Arvinas [E2].
Creation of spin-out company Amphista Therapeutics
The 2017 publication of the ligase-PROTAC-target crystal structure [R4] generated enormous investor interest. Amphista Therapeutics was consequently spun-out in December 2017 from the University of Dundee with co-founding venture capital investor Advent Life Sciences. In 2020, Amphista raised GBP6million Series A financing from co-investors Advent, the Scottish Investment Bank, the European Investment Fund and US-based BioMotiv [E3]. Amphista has created jobs for 15 employees (headcount: 15, FTEs:15) and is expected to grow to 25-30 in 2021. A General Partner of Advent Life Sciences explains:
”Advent invests in companies with first or best in class approaches…Amphista is an ideal example of the UK emulating some of the best US academic–industry partnerships. A world leading scientist who has pioneered an important new approach to the discovery of new medicines, working with an experienced Venture Firm to form a University spinout which then raises international finance to build an internationally recognised independent company operating within the UK. While there are some such companies in the UK, they are all too few compared to the US.” [E3].
Partnerships with Boehringer Ingelheim and others
In July 2016 Boehringer Ingelheim (BI) announced a significant (non-disclosed) investment in a collaboration to develop new classes of therapeutics based on PROTAC technology [E4]. The partnership, extended in 2018, created 12 (headcount: 12; FTEs: 12) research jobs in Dundee and approximately 30 (headcount 30: FTEs 30) at BI as part of a novel joint research team [E4, E5]. With an initial focus on oncology, the team progresses PROTACs against human targets suggested by BI, such as the BAF complex that is mutated in 20% of human cancers [E5]. Of the partnership, a Senior Vice President at BI said:
“Dundee University share our vision to transform cancer medicines with PROTACs. We extended our multi-million-pound investment in our alliance with Prof Ciulli to progress new PROTAC therapeutics and bring previously deemed undruggable targets within reach. Alessio Ciulli’s ground-breaking work in defining one of the first ever non-peptidic PROTAC degrader and mechanistic insights into how PROTACs work was instrumental in BI’s decision to collaborate with the University of Dundee” [E4].
The research has continued to attract interest, leading to GBP18million of collective investment since 2016 into Dundee from pharmaceutical companies BI, Nurix, Ono Pharma, Eisai and Almirall, and a total commercialisation income (royalties plus milestones) of GBP1,020,858 [E6].
Sale and open-access use of tools/reagents enabling PROTAC research and innovation
In 2018, Boehringer released Ciulli’s MZ1 on their Open Innovation OpnMe portal, as the first PROTAC freely-available to researchers [E7], with five Ciulli PROTACs now offered [E4]. Of 133 orders in 2019/2020, 45% came from private companies, with orders going to 25 countries [E4]. Two Dundee PROTACs are in the top 5 requested OPnMe molecules [E4]. Several biotech companies, including Tocris, ProbeChem, Medchemexpress and Cayman, market larger quantities of Ciulli’s PROTACs under license from the University of Dundee [E8] with current royalty income to the university of GBP200,000 [E6]. Life Sciences company Promega has also significantly benefited, as explained by their Group Leader of Research & Development:
“The early sharing with us by Alessio Ciulli of key PROTACs discovered in his lab, including but not limited to MZ1, was truly enabling to Promega. He provided fundamental tools to develop novel technologies to study targeted protein degradation and benchmark our efforts in this space, which is today one of the fastest-growing commercial areas for Promega” [E9].
5. Sources to corroborate the impact
[E1] Expert opinion piece citing Ciulli’s research [R3]: Scudellari, M. (2019) ‘Protein-slaying drugs could be the next blockbuster therapies’. Nature vol. 567, pp. 298-300. DOI: 10.1038/d41586-019-00879-3
[E2] Ciulli patents since 2016 and list of pharmaceutical industry patents citing Ciulli underpinning research R1-R6 and related patents.
[E3] Statement by General Partner of Advent Life Sciences Venture Capital Firm; plus article on Amphista Series A deal.
[E4] Testimonial by the Senior Vice President, Boehringer Ingelheim.
[E5] Evidence on the extension of the Boehringer Ingelheim partnership with Ciulli, and joint output evidencing the effectiveness of the partnership:
Press release: 2019, ‘Boehringer Ingelheim and University of Dundee Highlight Successful PROTAC Drug Discovery Program and Extend Their Ongoing Anti-Cancer Alliance’ Boehringer Ingelheim 11th June 2019.
Farnaby, W., Koegl, M., Roy, M. J., Whitworth, C., Diers, E., Trainor, N., Zollman, D., Steurer, S., Karolyi-Oezguer, J., Riedmueller, C., Gmaschitz, T., Wachter, J., Dank, C., Galant, M., Sharps, B., Rumpel, K., Traxler, E., Gerstberger, T., Schnitzer, R., Ciulli, A. (2019) ‘BAF complex vulnerabilities in cancer demonstrated via structure-based PROTAC design. Nature Chemical Biology, vo. 15, pp672-680. DOI: 10.1038/s41589-019-0294-6
[E6] Statement by Research Innovation Services at the University of Dundee verifying total investment, commercialisation income, and reagents sales income.
[E7] Article on the release of PROTAC MZ1 by Boehringer Ingleheim on their Open Innovation OpnMe portal, plus video about MZ1: 2019 ‘University of Dundee and Boehringer Ingelheim collaborate for free access of PROTAC compound on opnMe.com’ DiscoveryToday.com 9th Jan 2019; Video from Boehringer Ingelheim: 2019, ‘The Journey of MZ1, a BRD4 PROTAC, Dundee, Scotland’ Boehringer Ingelheim Jan 2019. Available at https://opnme.com/molecules/bet-mz-1
[E8] Web sales pages for University of Dundee PROTAC compounds sold by biotech companies:
Tocris Bioscience: MZ1 Cat.6154; AT1 Cat. 6356; CM11 Cat. 6416
Probechem: AT1 Cat. PC-35351
Medchemexpress: MZ1 Cat.HY-107425; AT1 Cat. HY-111433
Caymanchem: MZ1 Cat. 21622
Abcam: MZ1 Cat.ab230371
Lifesensors: MZ1 Cat. PC1001
[E9] Evidence of impact of Ciulli’s collaboration with Promega:
Statement by R&D Group Leader, Promega Corporation
Promega’s PROTAC drug discovery application webpage illustrating use of Ciulli’s MZ1 PROTAC: ‘Targeted Protein Degradation’ Promega. Available at https://bit.ly/2ZahIG4 (Accessed 16th Dec 2020)
Publication by Promega corroborating use of MZ1 in Promega technology development which cites R2-4: Riching, K., Mahan, S., Corona, C., McDougall, M., Vasta, J., Robers, M., Urh, M. and Daniels, D., (2018) Quantitative Live-Cell Kinetic Degradation and Mechanistic Profiling of PROTAC Mode of Action. ACS Chemical Biology, Vol. 13 No.9, pp.2758-2770. DOI: 10.1021/acschembio.8b00692