The next wave of scientific innovation - AstraZeneca

55 downloads 373 Views 4MB Size Report
Guangzhou Medical College, China ...... hits, leads and drug candidates that will be safe and ... To complement our Hit
The next wave of scientific innovation Innovative Medicines & Early Development Biotech Unit 2015 – a year in review

Oncology combination therapies AstraZeneca is investigating combinations of biologic and small-molecule therapies for the treatment of cancer. These combinations target the tumour directly and some help boost the body’s own immune system to induce tumour cell death.

Contents

Introduction 2 The next wave of scientific innovation An introduction from Mene Pangalos 4 IMED 2015 in numbers 6 The story of osimertinib (or AZD9291) Our 5R framework in action

Biologics in the treatment of asthma

The next wave of scientific innovation 1

An environment where science thrives

An environment where science thrives 92 People: Inspiring great scientists 96 Our strategic science centres 98 Building our future in Cambridge 100 Case study: The next wave of innovation in DNA damage response 104 Our reputation for scientific leadership 108 Preparing for the future with our ‘IMED Futures’ teams

Collaborating for science innovation

CGI image of AstraZeneca’s new Global R&D Centre and Corporate Headquarters, Cambridge, UK

Collaborating for science innovation 82 Case study: Partnering to develop new medicines for neurodegenerative diseases 84 Three science units with one shared goal 86 Partnering to redefine the future of drug discovery 90 Case study: Partnering to develop the next generation of antisense-based therapeutics

IMED functions

IMED functions 46 Discovery Sciences 52 Case study: The technology supporting our science – Acoustic mass spectrometer 54 Drug Safety & Metabolism 62 Personalised Healthcare & Biomarkers 72 Early Clinical Development 80 Shaping drug development in Asia

Therapy area progress

Therapy area progress 10 Oncology iMed 18 Case study: The technology supporting our science – Confocal Microscope 20 Respiratory, Inflammation & Autoimmunity iMed 30 Case study: PT010 triple combination speeds into Phase III 32 Cardiovascular & Metabolic Diseases iMed 40 Neuroscience iMed

Introduction

Delivering the next wave of scientific innovation

The next wave of scientific innovation An introduction from Mene Pangalos In the last 12 months, we have continued to demonstrate the strength of our science, setting new records to deliver lifechanging medicines to patients around the world. All of this has been made possible through the dedication and commitment of our amazing people, and their relentless passion for science and innovation.

2015 also saw further progress in our pioneering approach to open innovation. Thanks to the outstanding work of a small team of people from our Strategic Partnering & Alliances group, working alongside colleagues from our iMeds and Functions, we have built a virtual pipeline supporting more than 20 clinical and 80 pre-clinical studies. Since the launch of our IMED Open Innovation portal in October 2014, we have seen >10,000 visits and reviewed >350 proposals for new drug projects. It’s a great example of how our scientists are being entrepreneurial, creating an environment that is open for collaboration and challenging conventional thinking.

In the last quarter, our organisation entered into an exclusive agreement with the Wallenberg Centre for Protein Research to identify new targets for disease research in the groundbreaking area of the Secretome – research into all proteins that are secreted by a cell or that are exposed to the outside of the cell from within the cell membrane. The momentum continued right up to the end of the year. In December, our IMED scientists played a pivotal role, delivering the agreement to acquire a majority equity stake in Acerta Pharma, which gives us access to acalabrutinib (ACP-196), a potential best-in-class, smallmolecule oral BTK inhibitor.

Left Mene Pangalos, Executive Vice President, IMED Biotech Unit Above CGI images of AstraZeneca’s new Global R&D Centre and Corporate Headquarters, Cambridge, UK

2 ©AstraZeneca 2016

Although not possible to capture all of the details of the incredible contributions of our people across the IMED Biotech Unit in 2015, I hope this report gives you a flavour of the work we have done and the role we have played in helping to ensure AstraZeneca continues to push the boundaries of science.

Mene Pangalos Executive Vice President, IMED Biotech Unit

The next wave of scientific innovation 3

An environment where science thrives

Throughout the year, our teams continued to look to the future – exploring and investing in the next wave of scientific innovation. We supported recommendations for investment for our IMED Futures teams to explore emerging technologies and innovations that have the potential to redefine the future of healthcare. We invested in organs-on-a-chip technology that could one day become an entire human body on a chip, bringing us closer to our goal of reducing, refining and replacing the use of animals in research. We also made significant discovery investments to enhance the use of CRISPR precision DNA-editing technologies across our discovery platforms, and we are looking forward to further understanding its application as a therapeutic option in our main therapy areas.

Collaborating for science innovation

Another personal highlight is our progress in personalised healthcare (PHC), developing targeted treatments matched using diagnostics to the patients most likely to benefit, which has been rapid by industry standards. In 2015, we launched seven diagnostic tests and signed 13 diagnostic partnerships. These achievements position AstraZeneca as a leading company in personalised healthcare amongst our peers – a tremendous accomplishment since the formation of our Personalised Healthcare & Biomarkers group in 2011. We also announced a collaboration with the Montreal Heart Institute (MHI) to search the genomes of up to 80,000 patients for genes associated with cardiovascular diseases and diabetes to support the discovery of new targeted treatments for these serious conditions. The AstraZeneca Senior Executive Team further demonstrated the company’s commitment in this area at the end of 2015 by agreeing a broader genomics strategy across the spectrum of our discovery and development activities, something we look forward to sharing more about in 2016.

IMED functions

“In the last 12 months, we have continued to demonstrate the strength of our science, setting new records to deliver life-changing medicines to patients around the world.”

Secondly, our Respiratory, Inflammation and Autoimmune team achieved a Phase III investment decision for PT010, our triple combination inhaler for the treatment of patients with COPD. The combination builds on the previously successful Phase III outcome of our dual-LAMA/LABA combination

The quality of our science was also demonstrated with our teams setting new records for scientific publications. Our transformation in the last five years is nothing short of exceptional, moving from a single high-impact publication in 2010 to a record of 29 high-impact publications in 2015. This not only reflects the quality of the research conducted in our laboratories, but also demonstrates how our sciencedriven culture is helping to build the scientific reputation of AstraZeneca.

Therapy area progress

During the year, our teams progressed a number of important projects within our main therapy areas, helping to further strengthen the AstraZeneca pipeline. While it is difficult to pick out highlights, I will call out three key progressions:

Firstly, in oncology, where we saw the approval of osimertinib (AZD9291) and the accompanying companion diagnostic in the US in lung cancer. This is one of the fastest, if not the fastest, drug approvals from first-time-in-human to launch in less than three years. Adding a positive CHMP opinion towards the end of the year rounded off an incredible achievement by our teams, of which I am incredibly proud.

Finally, in CVMD, it was great to see the anti-microRNA from our Regulus partnership, AZD4076 (anti-miR103/107) for nonalcoholic steatohepatitis, reach the clinic. Oligonucleotides play an increasingly important role in our research portfolio, seeking to target novel proteins at the desired tissue more effectively, so these were important achievements for our team.

Introduction

2015 has been a remarkable year for our company, and for the IMED Biotech Unit.

(PT003), and will also include the anti-inflammatory inhaled corticosteroid budesonide. Our proprietary porous particle co-suspension technology in the pMDI, developed by our colleagues at Pearl Therapeutics, allows aerodynamically efficient drug delivery.

IMED Biotech Unit by numbers in 2015 Around

60

Over

120

60

12

clinical project combinations in oncology

major collaborations

post-docs

Phase I and Phase II starts

90%

29

high-impact publications

projects with Personalised Healthcare approach

Over

29

$1bn investment in scientific research

clinical projects

7

2

2500

positive Phase III investment decisions

Nearly

$50m generated through Open Innovation

4 ©AstraZeneca 2016

companion diagnostics launched

people with a passion for science

Almost

450

peer-reviewed publications

The next wave of scientific innovation 5

The story of osimertinib (or AZD9291) Our 5R framework in action

AZD9291 – or osimertinib as it is now known – received Food and Drug Administration (FDA) approval in November.

The stats Positive data on osimertinib in first-line EGFR mutated lung cancer at the American Society of Clinical Oncology (ASCO) meeting 2015: – 81% patients on a once-daily dose of osimertinib were progression free at nine months – Overall response rate 73% – Longest duration of response was ongoing at 13.8 months at the time of data cut-off Therapy area progress

1.59 million people die of lung cancer every year, one of the biggest cancer killers in the world. Around 80-85% of lung cancers are non-small cell (NSCLC) and its five-year survival rate is less than 10%. Osimertinib was approved in the US for the treatment of patients with metastatic EGFRm T790M non-small cell lung cancer who have received prior EGFR-TKI therapy.

The T790M ‘gatekeeper’ mutation is prevalent in approximately two-thirds of cases of EGFRm advanced NSCLC. This was our biological target, and data from our clinical trials gave us confidence that osimertinib would have a clinically meaningful benefit and address an area of high unmet medical need.

“The science in our labs that generated AZD9291 has been absolutely fantastic. I still remember the first time our scientists showed me our molecules binding to the T790M mutated receptor in 2010. The understanding of that science, the quality of the chemistry that we were doing, the biology, to enable us to progress the molecule so quickly through the research phase and ultimately get the candidate out in March 2013 was very, very impressive. This is something that I think all our scientists can be extremely proud of.” Mene Pangalos, Executive Vice President, IMED Biotech Unit

Introduction

The story of osimertinib or AZD9291 is a story of the fastest ever drug to make it from discovery to market. And it all began right here with our IMED Biotech team, living our values to follow the science, and challenge everything.

“I arrived at AstraZeneca in September 2010, and one of the first jobs that I had to do was go through and review the portfolio to rank and prioritise projects. AZD9291 was in discovery at the time and I think it’s helpful to go back to the origins of thinking ‘why would we want a drug like this?’ because at the time it wasn't as obvious as it is now that you have the data. I think you have to recognise that the scientists that came forward with this idea had to struggle against some prevailing wisdom that this might not be a useful thing to do.” Susan Galbraith, Head of Oncology iMed

Our 5R framework in action Quality – not quantity

designed to overcome a common resistance mechanism, ‘gatekeeper’ mutation T790M



IMED functions

Right target

Osimertinib was one of the first examples where we applied the 5R approach, when it was identified as a ‘must-win’ project.

Right tissue good bioavailability and widely distributed in tissues, potential objective response rate of 66%

Right safety good tolerability profile, minimised hyperglycaemia risk and unwanted activity on the receptor which causes rash and diarrhoea

Right patient

Collaborating for science innovation



companion diagnostic – cobas® EGFR Mutation Test v2 for the detection of T790M mutations in both tumour tissue and blood – developed in partnership with Roche Molecular Systems

Right commercial opportunity defining the value, understand more quickly and deeply the patient subgroups and future viability



Plus… Right culture team speed and flexibility to capitalise on the opportunity, and drive innovation in every aspect of how we discover and develop new therapies

6 ©AstraZeneca 2016

The next wave of scientific innovation 7

An environment where science thrives



Innovation in the clinic A clear vision

Needless to say, screening hundreds of lung cancer patients to find the few with the correct genetic driver is slow and expensive, challenging for physicians, and frustrating for the patients – who have a lower probability of having the correct genetic driver for the single targeted drug being tested.

This approach puts the patient at the centre of the trial, offering more drug options for the patient through a single trial process and a better chance of getting on the trial, while allowing us to test the drugs more quickly and effectively. Basket trials speed up the discovery of new medicines making it possible to offer better medicines, to more patients, more quickly.

13

Genetic drivers of disease

ATM

Drug

AZD6738

5

14

3

38

15

30

7

4

13

MET

PIK3CA pathway

LKB1 or TSC1/2

FGF2/3

KRAS or NF1

EGFRm+

PIK3CA pathway

LKB1 or TSC1/2

FGF2/3

KRAS or NF1

savolitinib

AZD5363

AZD2014

AZD4547

Selumetinib

Osimertinib or Gefitinib

AZD5363

AZD2014

AZD4547

Selumetinib

5

Figure highlighting % of patients with different genetic drivers of lung cancer and the variety of drugs included in the basket trial to attack each of those specific genetic drivers.

What next for osimertinib? Our teams continue to keep driving and keep looking for novel and next-generation strategies to benefit patients – from exploring potential benefits in other settings, combination therapies and first-line therapy. We are also starting to see that the same type of science and the same type of approach can be used in other patients with other diseases beyond oncology. “The future is very exciting and osimertinib highlights a key milestone in building our oncology pipeline. We’ve launched olaparib, we have gefitinib, now osimertinib, and we are keeping our eyes to the big milestone, launching our immuno-oncology pipeline. This is a great time to be in oncology in AstraZeneca.” Mene Pangalos, Executive Vice President, IMED Biotech Unit

The importance of collaboration The companion diagnostic for osimertinib was developed in partnership with Roche Molecular Systems (RMS). For patients who may not be able to provide a biopsy or whose test result based on a tissue sample is unknown, then circulating tumour DNA (ctDNA) offers a solution in terms of a source of DNA that can be analysed for mutations. In this case, the diagnostic approach developed with RMS was especially important, as assay development for the T790M mutation that we were looking to detect is challenging because of the sequence surrounding that mutation. Our early exploratory work enabled us to find the test that could detect the mutation in plasma as well as tissue.

“To move so fast, you have to start from a very solid foundation. In this case the solid foundation was really, really excellent science. There was scientific purpose, there was excellence by design if you will. The drug was designed to do a job, and it did it.” Flavia Borellini, Global Medicine Leader AZD9291, Global Medicines Development

8 ©AstraZeneca 2016

The next wave of scientific innovation 9

An environment where science thrives

“It was just amazing to sit in a room and actually see a slide of a patient before and a patient after six weeks of treatment and the tumour’s shrunk by 60%. It just doesn’t get any better in terms of the science that we do here. For me it was incredibly rewarding. Susan Galbraith came into the office and I’ve never seen her so excited, she was practically dancing. It was amazing!” Ray Finlay, Medicinal Chemistry, IMED Biotech Unit

% of population

Collaborating for science innovation

“From the first time this molecule went into patients we were seeing a clinical response and that was just fantastic news for patients, for the team who worked on the molecule and for the organisation as a whole.” Mark Anderton, Discovery Toxicologist Drug Safety & Metabolism, IMED Biotech Unit

Squamous NSCLC

Adenocarcinoma NSCLC

IMED functions

An emerging solution to these challenges, which leverages our broad portfolio in lung cancer, are ‘basket’ trials. These are trials that don’t test a single drug, but a ‘basket’ of drugs, each one targeting a specific molecular or genetic driver of disease. Rather than screening for a single genetic driver in a patient population, we can then screen patients for drivers of their disease and match the right patient to the right drug in the ‘basket’.

Patient cohort on trial

Therapy area progress

“In a project, you keep on making compounds, testing compounds, really until you’re absolutely sure you have a high-quality compound, clinical candidate. We had some early compounds that looked interesting. Unfortunately, when we looked at them in more detail we thought some of them had an issue with hyperglycaemia, which we believe is caused by off-target inhibition of the insulin receptor. As a chemistry team we were able to design out that activity with AZD9291, so AZD9291 came through and didn’t have that liability – this is something that we’re really proud of as a chemistry team.” Richard Ward, Principal Scientist Chemistry, IMED Biotech Unit

Traditional clinical trials use a single drug for a given patient population. If that population is small (for example, genetically defined as 1% of the population), we would need to screen 100 patients to find the one patient that will benefit from the targeted drug.

Basket trial

Introduction

“The early project team were a fantastic group of people to work with. I think what really helped was we had a really clear vision of what we wanted to do. We’d done a lot of discussion and a lot of consultation about what the profile of the molecule needed to be and we’d expanded it from its original vision and then we were really clear. The molecule needed to look like this. It needed to have these properties and this kind of potency, not this kind of toxicity. I think that really helps and the team really gelled. Initially we weren’t under the spotlight that AZD9291 is today. It was a bit of a slow burner if you like, but we delivered it very, very quickly.” Teresa Klinowska, Early Project Leader Oncology iMed, IMED Biotech Unit

Our increasing ability to identify the molecular or genetic mechanisms that drive cancer, such as non-small cell lung cancer (NSCLC), has allowed us to recognise and select defined patient subgroups where an individual’s cancer (disease segmentation) is identified by the molecular or genetic driver. However, this brings new challenges in drug development as patient subpopulations consequently become smaller.

Oncology iMed Our vision is clear. To help patients by redefining the cancer-treatment paradigm, with the aim of bringing six new cancer medicines to patients between 2013 and 2020. A broad pipeline of next-generation medicines is focused principally on four disease areas – breast, ovarian, lung and haematological cancers.

10 ©AstraZeneca 2016

The next wave of scientific innovation 11

Oncology iMed

Susan Galbraith, VP Oncology iMed

Opposite Cancer cells Top Susan Galbraith, VP Oncology iMed

The iMed saw significant progress in its early discovery phase and clinical phase portfolio. In addition to nominating three new candidate drugs, we started

Oncology also provided extensive support for the late-stage and Phase III pipeline: Osimertinib: Provided data supporting the potential for osimertinib in early-stage disease, supported circulating tumour DNA testing and generated exciting efficacy data in leptomeningeal disease. Olaparib: Provided scientific foundation for expanding the use of olaparib beyond germline BRCA for Solo 2 (ovarian cancer) trial and gastric trials as well as to support the Phase III investment decision in prostate cancer. Faslodex: Delivered data showing activity of Faslodex in tumours with ESR1 mutations.

Therapy area progress

Beyond these exciting data, there were a total of 11 oral and 19 poster presentations with iMed authors at ASCO 2015.

clinical development of an ATM inhibitor (AZD0156) and an aurora kinase inhibitor (AZD2811). We moved three projects into Phase II clinical trials and made significant progress with our existing Phase II assets. Introduction

“We have made a huge amount of progress in a short time while moving to our new location in Cambridge this year, including supporting Phase III investment decisions for osimertinib in the adjuvant setting, and expansion in leptomeningeal disease, and olaparib in prostate cancer. We also achieved three candidate drug investment decisions and two compounds entering first time in human trials. There was great progress in the Discovery phase, setting us up for exciting candidate drug progressions in 2016 and 2017. We now have over half our UK-based staff in Cambridge, and I would like to thank everyone involved in making this transition successful.”

The approval of osimertinib towards the end of the year was a key highlight, just 32 months after first-dosing in patients. At the American Society of Clinical Oncology (ASCO) meeting 2015, AstraZeneca presented positive data on osimertinib in first-line EGFR mutated lung cancer. Data showed that 81% of patients on a once-daily dose of osimertinib were progression-free at nine months, with an overall response rate of 73%. The longest duration of response was ongoing at 13.8 months at the time of data cut-off.

Highlights

Opportunities with Starpharma and Heptares Therapeutics. For Starpharma, this was for the use of their dendrimer drug delivery technology in the development and commercialisation of one of our oncology compounds with the potential to add additional compounds later. Heptares Therapeutics – we have gained exclusive global rights to develop, manufacture and commercialise HTL-1071 (now AZD4635), an adenosine A2A receptor antagonist.

Demonstrate progress in at least two scientific areas based on external collaboration.

22 papers derived from collaborations published /accepted in 2015. This included several publications in impactful journals, including on PI3K inhibition in Cancer Cell and on blockade of AKT and MEK in Ras-driven tumours in Clinical Cancer Research

Deliver new portfolio opportunities in an Emerging or Asian Oncology Market.

Collaboration with China team on osimertinib; three Korean master agreements; launch of Taiwan translation fellowship programme; Koc University (Turkey) studentship for target validation.

Build a credible small-molecule drug discovery effort around immunooncology targets.

A strategic push to explore possibilities for small-molecule drugs and relevant targets in the exciting immuno-oncology arena with the idea of integrating with and complementing the checkpoint antibodies and other biologics coming from our MedImmune colleagues. We developed key capabilities and hired staff with strong backgrounds in immunology. We focused on factors that promote the immunosuppressive microenvironment that exists in many tumours. We now have two ongoing clinical trials with antagonists of STAT3 signalling and CXCR2, both combined with durvalumab and we recently licensed a clinical phase Adenosine A2a antagonist (AZD4635).

Refresh our early-stage discovery strategy.

Focused effort into three main areas – oncogenic drivers and resistance mechanisms, DNA damage response biology and finally immuno-oncology.

To deliver a comprehensive solution for patients with EGFR-driven non-small cell lung cancer.

A drug combination strategy to drive deeper and more durable responses and have ongoing clinical studies combining our EGFR inhibitors with inhibitors of both MEK and cMet, as well as a collaboration with Incyte to explore JAKi, all pathways that have been shown pre-clinically and/or clinically to contribute to drug resistance. Finally, while osimertinib represents an important solution for patients with T790M resistant disease we are committed to exploring resistance mechanisms that arise in patients and initiating efforts to address those events.

The next wave of scientific innovation 13

An environment where science thrives

Close two licensing deals.

Collaborating for science innovation

We delivered

IMED functions

12 ©AstraZeneca 2016

We set out to

Highlights We delivered

Enhance AstraZeneca’s capability to perform Next Generation Sequencing programmes.

Augmented laboratory and computational Next Generation Sequencing (NGS) capabilities, which increased throughput and delivery for oncology projects. NGS support of pre-clinical and clinical programmes resulted in programme line-of-sight, targets, PHC, biomarker assays, CDx development, model characterisation, genetic drivers, and mechanisms of resistance. As we work to build an integrated sample-to-answer workflow, Oncology NGS Lab and Production Informatics’ efforts have matured from methods development to methods optimisation and scale-up. These new tools will enable biologists to more seamlessly integrate sample and patient data to explore and understand genotype and phenotype and begin to deliver on the promise of precision medicine.

Build our Phase II pipeline.

– Data sets that showed AZD1775 (Wee1 inhibitor) has activity in combination with platinum-based chemotherapy in ovarian cancer. Data from a single-centre study in Platinum-resistant and refractory disease demonstrated a 41% response rate and was presented at the ASCO meeting. Importantly, combination dosing of AZD1775 with both durvalumab and olaparib was initiated in 2015

– Three compounds into Phase II clinical trials. AZD9150 is a STAT3 antisense oligonucleotide, which started a Phase II trial dosing in combination with anti-PD-L1 durvalumab. The trial also allows us to evaluate the combination of durvalumab with AZD5069, a small-molecule inhibitor of the chemokine CXCR2. The third compound was AZD3759, an EGFR inhibitor designed to have increased brain penetration. AZD3759 met the predetermined PoM criteria

An approved companion diagnostic for osimertinib, and worked to identify and characterise mechanisms of clinical resistance. In addition, the team led work to support potential for olaparib beyond germline BRCA in ovarian, gastric and prostate cancers.

Create the Cambridge Cancer Science Symposium with MedImmune.

A successful event with 286 attendees, 12 topics, 75 talks, 136 posters, 17 academic institutes that has encouraged early project openness and identified many immunooncology and combination approaches.

Olaparib In 2015, the Phase III investment decision was made for olaparib in metastatic castrate resistant prostate cancer (mCRPC). There is a high unmet need and an opportunity to advance standard of care (SOC) with personalised medicine. Prostate is the most common cancer in men and the sixth leading cause of cancer death among men. New hormonal agents (abiraterone and enzalutamide) and taxanes (docetaxel and cabazitaxel) are SOC. Although the heterogeneity of mCRPC is well recognised; treatment to date has not been driven by companion diagnostics (CDx). In 2015, we have generated limited but very encouraging data

14 ©AstraZeneca 2016

from TOPARP A (biomarker +ve N=16) with an 88% response rate and mean progression-free survival of 9.8 months. Studies are ongoing with the new data expected towards the end of 2016. FDA provided useful insights, which will support our development plan and innovative trial design given the rarity of the patient population. In addition, the translational science team have provided support for broadening the opportunity for olaparib beyond germline BRCA by developing the SOLO2 (confirmatory Phase III trial in second-line ovarian cancer) for somatic tumour-based BRCA testing plan, and by running a successful concordance study for other mutations in genes involved in Homologous Recombination Repair (HRR) with Myriad and Foundation Medicine.

Dan Stetson was a major contributor to the introduction of Next Generation Sequencing and has continued to drive the development of methodology for projects and novel technologies. Dan led the validation of the Illumina platform for NGS.

As a member of the KRAS team, Sarah Ross provided a significant contribution to the strategy for candidate nomination. Sarah saved significant investment on an HTS through detailed biochemistry. This was achieved in a year when Sarah moved to Cambridge, has taken on some line accountability and still delivers from the bench.

Paul Lyne completed a phenomenal achievement this year – successfully leading all three projects that delivered Candidate Drugs for the iMed. The AZD4785 (Kras), AZD4635 (A2aR) and AZD4205 (JAK1) projects all progressed into the Pre-clinical stage of development in 2015. These novel targets required the team to build creative development plans. In addition, Paul has made major contributions to the development of the next generation of immune-modulatory agents through the delivery of the Phase II starts for AZD9150 (STAT3) and AZD5069 (CXCR2) as well as working with MedImmune to coordinate the portfolio of small-molecule-largemolecule combination studies.

The next wave of scientific innovation 15

An environment where science thrives

Powering the Phase III pipeline

People spotlight

Collaborating for science innovation

Support the late stage and Phase III pipeline.

AZD9291 ADAURA study Development of AZD9291 for early stage EGFRm disease is a key component of the AstraZeneca lung cancer strategy. The opportunity in adjuvant EGFRm NSCLC has many compelling aspects. Firstly, there is an unmet need to address the high relapse rate and impact long-term survival through long treatment duration. This is possible due to AZD9291 tolerability profile. AstraZeneca is ideally placed to strengthen its scientific leadership in NSCLC and personalised medicine as the adjuvant population is expected to increase, especially in the US and Japan, due to improved screening and early detection. We are now embracing the next wave of scientific innovation and enrolling EGFRm patients in the adjuvant setting to meet recruitment targets to development this new modality.

IMED functions

– Encouraging response rate data with AZD5363 (AKT) monotherapy in AKT1 mutant breast cancer; to drive increased durability of response the first patients have been dosed with a combination of AZD5363 and Faslodex

The primary objective was to assess the safety and tolerability of osimertinib in patients with LM. All 13 patients were Asian with adenocarcinoma. Three of eight patients had improved neurological exam per investigator; of five patients with normal neurological exam at baseline, four had no change. Eight patients are continuing treatment with osimertinib beyond four months. One patient with neurological improvement was

This initial data has been very well received at global and US Advisory Boards and we are now enrolling a larger expansion cohort.

Therapy area progress

– Full recruitment of the Phase II trial for savolitinib (AZD6094; a cMet inhibitor) in papillary renal cell carcinoma and preliminary data from the TATTON trial was presented at the ASCO meeting, which supports combination therapy in patients with a cMet amplification who lack the T790M secondary mutation

There is no established effective treatment for LM disease. Different treatment approaches are used such as radiation, systemic or intrathecal chemotherapy with limited success. The overall survival is 7-14 months with LM from EGFRm NSCLC previously treated with an EFGR tyrosine kinase inhibitor (EGFR-TKI). However, through collaboration with our Innovation Center in China, we have established a preclinical rationale for osimertinib activity in CNS disease and low incidence of progression in the brain among patients without brain metastases suggests potential for CNS control. Addressing an important unmet need, the opportunity for differentiation was highlighted at Portfolio Review in June 2015 to support the case for an expansion study in Q4.

a 62-year-old Korean male, diagnosed with advanced nonsmall cell lung cancer (Ex19del, T790M) in March 2012 with most recent progression due to LM spread in March 2015. Prior therapy included multiple courses of chemotherapy, an EGFR-TKI and WBRT in 2014. He presented with right lower extremity weakness, headache and dyspnea. He started with 160 mg AZD9291 in April 2015. The LM response was ongoing from week six, as was his extracranial disease. He showed improved motor function while on treatment, progressing from wheelchair bound to being able to walk with cane.

Introduction

We set out to

Osimertinib The case was also successfully made to expand the osimertinib cohort in a CNS disease study, to characterise and explore label inclusion for leptomeningeal disease. Leptomeningeal Metastasis (LM) is a devastating complication of NSCLC and has been reported in 4–15% of NSCLC patients, resulting in a very poor prognosis. An increased risk of CNS involvement has been reported among patients with EGFRm NSCLC, in particular those treated with a first-generation EGFR-TKI.

Oncology iMed small-molecule pipeline end of 2015

Phase I

Phase II

Phase III / Launch

AZD4635/A2aR

AZD4547 / FGFR

AZD5363 / AKT

Selumetinib / MEK

AZD4785/KRAS ASO

AZD8186 / PI3Kb

AZD2014 / TOR

AZD9291 / EGFR

AZD4205/JAK1

AZD6094 / cMet

AZD9496 / SERD

AZD9150 / STAT3 AZD5069/ CXCR2

AZD0156 / ATM

AZD3759/EGFR-BBB

Schug Z, Peck B, Zhang Q, Jones DT, Grosskurth S, Alam I, Smethurst E, Mason S, Byth K, McGarry L, James D, Shanks E, Kalna G, Saunders B, Jiang M, Howell M, Lassailly F, Thin MZ, Spencer-Dene B, Stamp G, Harris A, Abogaye E, Critchlow S, Wakelam M, Schulze A, Gottlieb E

Cancer Cell

Feedback suppression of PI3Ka signalling in PTEN-mutated tumours is relieved by selective inhibition of PI3Kβ

Schwartz S, Wongvipat J, Trigwell CB, Hancox U, Carver BS, Rodrik-Outmezguine V, Will M, Yellen P, de Stanchina E, Baselga J, Scher HI, Barry ST, Sawyers CL, Chandarlapaty S, Rosen N

Nature Reviews Drug Discovery

An analysis of the attrition of drug candidates from four major pharmaceutical companies

Waring MJ, Arrowsmith J, Leach AR, Leeson PD, Mandrel S, Owen RM, Pairaudeau G, Pennie WD, Pickett SD, Wang J, Wallace O, Weir A

Nature

Patient-centric trials for therapeutic development in precision oncology

Biankin AV, Piantadosi S, Hollingsworth SJ

Nature Medicine

Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harbouring EGFR T790M

Thress KS, Paweletz CP, Felip E, Cho BC, Stetson D, Dougherty B, Lai Z, Markovets A, Vivancos A, Kuang Y, Ercan D, Matthews SE, Cantarini M, Barrett JC, Janne P, Oxnard G

Journal of Clinical Oncology

Randomized, double-blind Phase II trial with prospective classification by ATM protein level to evaluate the efficacy and tolerability of olaparib plus paclitaxel in patients with recurrent or metastatic gastric cancer

Bang YJ, Im SA, Lee KW, Cho JY, Song EK, Lee KH, Kim YH, Park JO, Chun HG, Zang DY, Fielding A, Rowbottom J, Hodgson D, O'Connor MJ, Yin X, Kim WH

Samsung Medical Centre, Seoul, South Korea

Oregon Health Sciences University, Portland, Oregon and the Leukemia and Lymphoma Society, New York, US

Bind Therapeutics, Boston, US

Gastric Cancer Alliance. Basket study with selumetinib, savolitinib, AZD1775, AZD5363, and AZD2014 plus paclitaxel in second-line Gastric Cancer, with biopsies.

Test primary AML samples with biomarker/genetic association follow-up.

Development of AZD2811 nanoparticle aurora B kinase inhibitor. Nanoparticle approach provides a slow release profile so a single infusion will give multiple-day efficacious cover of the target and delivered improved therapeutic index in pre-clinical models.

Nature Reviews Cancer

MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road

Caunt CJ, Sale MJ, Smith PD, Cook SJ

Nature Reviews Molecular Cell Biology

Targeting the DNA damage response in cancer

O’Connor MJ

Cell Metabolism

Leptin, BMI and metabolic gene expression signature are associated with clinical outcome to VEGF inhibition in colorectal cancer

Pommier AJC, Farren M, Patel B, Wappet M, Michopoulos F, Smith NR, Kendrew J, Frith J, Hubby R, Eberlein C, Campbell H, Womack C, Smith PD, Robertson J, Morgan S, Critchlow SE, Barry ST

Vall d’Hebron Institute of Oncology, Barcelona, Spain

Driving open innovation through clinical partnerships with 16 French Centres. Effective working across IMED, GMD, MedI and AstraZeneca France.

Genetic Determinants of Wee1 and Parp inhibitor sensitivity. Identify genetic backgrounds that correlate with sensitivity to olaparib and AZD1775 and the effectiveness of the combination.

An environment where science thrives

Institut National du Cancer, Luxembourg City, Luxembourg

Collaborating for science innovation

Acetyl-coA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress

IMED functions

Cancer Cell

AZD1775 / Wee1

Key Oncology iMed collaborations in 2015

16 ©AstraZeneca 2016

Authors

Olaparib / PARP

AZD8835 / PI3Ka

AZD2811 / AUR-N

Title

Therapy area progress

AZD6738 / ATR

Publication

Introduction

Pre-clinical

Key Oncology iMed publications in 2015

The next wave of scientific innovation 17

Case study

Behind the scenes

Automated confocal microscope Our technique of choice for high resolution microscopy

The technology supporting our science

The stats

– High throughput, fast, automated confocal imaging of live and fixed cells and tissue

– Faster than any other automated microscope that we had previously. For example, it can image a 384 well plate in three colours (one field of view) in around five minutes. Compare this to other widefield microscopes that we have, which can take ten times longer to image the same plate.

– Imaging with spatial cellular resolution (to monitor cellular trafficking, for example) – Imaging in multiple formats from slides to 1536 MTP plates

"This instrument promises new capability with improved assay quality at fast acquisition rates and will enable Discovery Sciences to expand their assay portfolio to support all of the IMED Biotech Unit."

– Enabling capabilities that we didn’t have before – confocal imaging at higher throughput. The only true confocal platform that we had previously was a stand-alone microscope which was only capable of processing single samples and so imaging a 384 well plate could take a scientist several days, now this can be achieved in as little as five minutes.

Case study

The facts

The Automated confocal microscope (CV7000) is an automated confocal fluorescence microscope that can acquire images from live or fixed cells and tissue samples. Confocal microscopy is an imaging technique widely used for increasing cellular spatial resolution by eliminating out-of-focus light through the use of pinholes placed at the confocal plane of the lens. Confocal microscopy has become an essential tool for the life sciences and is the technique of choice for high resolution microscopy.

The scientist perspective “The use of more physiological, cellular and tissue models including primary cells, stem cells and multicellular systems in preclinical drug discovery is now commonplace. Such models are often 3D in nature and therefore cannot be effectively imaged by traditional wide-field systems. In addition, AstraZeneca has invested heavily in phenotypic screening initiatives and precise genome editing. Having access to powerful detection microscopes that can detect subtle cellular phenotypic changes is critical to realising the full potential of such investments. We evaluated current and future project demand and it was clear that there were many projects spanning our R&D functions that could benefit from the purchase of the CV7000. This instrument promises new capability with improved assay quality at fast acquisition rates and will enable Discovery Sciences to expand their assay portfolio to support all of the IMED Biotech Unit. The ability to image and extract data from more complex cellular models with improved physiological relevance early in drug discovery will speed up the discovery process and reduce attrition. The microscope was installed at the latter end of 2015 and we have already started to use the system to impact projects where cellular spatial resolution is a requirement and for use with thicker tissue specimens. We are open to collaboration and would encourage interested parties to get in touch if they are interested in getting access to our microscopes.” Samantha Peel, Senior Research Scientist, Discovery Sciences

Confocal imaging of a 384 well plate could take a scientist several days, now this can be achieved in as little as five minutes 18 ©AstraZeneca 2016

The next wave of scientific innovation 19

Respiratory, Inflammation & Autoimmunity iMed AstraZeneca holds a unique position in respiratory disease, including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF), with a range of differentiated, potential medicines in development by leveraging novel combinations, biologics and devices. The pipeline also has a number of promising assets in inflammatory and autoimmune diseases within areas such as psoriasis, psoriatic arthritis, gout, systemic lupus and rheumatoid arthritis.

20 ©AstraZeneca 2016

The next wave of scientific innovation 21

Respiratory, Inflammation & Autoimmunity iMed Introduction

“2015 has been a transformative year where we have become fully staffed with the optimal scientific profile. The right mix of scientists is now breathing science and working on the RIA iMed portfolio, which aims to transform the lives of patients with respiratory, inflammatory and autoimmune diseases.” Maarten Kraan, VP RIA iMed

Therapy area progress

Our third breakthrough was discovering a vastly improved approach to access crystalline material in the early phase of inhaled delivery projects. This allows crystallisation conditions to be identified in a reproducible and controlled way, and consume only small amounts of compound. Multiple studies can be automated and processed in parallel, effectively removing one of the major bottlenecks in inhaled drug discovery projects.

An environment where science thrives

By polarising human alveolar macrophages ex-vivo, we have been able to identify unique COPD-specific gene signatures. The identification of novel gene signatures for polarisation status has led to new understanding of potential disease-relevant functions of alveolar macrophages in COPD. In addition, the experiments uncovered a remarkable cell plasticity independent of disease severity, opening up the potential for new therapeutic strategies targeting alveolar macrophages polarisation in COPD.

Collaborating for science innovation

Our ambition is to drive the scientific excellence agenda and lead the development of game-changing, inhaled, immuno-modulatory treatments for asthma and COPD patients. All core functions work together to provide the necessary scientific innovation, patient insight and technology to achieve that goal. Some significant, industryleading breakthroughs in 2015 included: characterisation of gene signatures for subsets of alveolar macrophages towards modulation of innate immunity in COPD; first ever measurement of in vivo lung receptor occupancy and early exploration of crystallisation conditions to fundamentally change inhaled drug dose estimation.

We have created a new paradigm in inhalation drug discovery. Without the means to assess the relationship between local tissue exposure and unbound pharmacologically active drug after inhalation, it has been notoriously difficult to establish solid PK/PD understanding for inhaled drugs. Our Drug Metabolism and Pharmacokinetics group has developed a new methodology to determine receptor occupancy in the lung, thus providing an elegant solution for describing PK/PD relationships. This was accompanied by a novel and sophisticated mathematical model, which gave input into drug design strategies and a way to better predict clinical outcome.

Above Maarten Kraan, VP RIA iMed

22 ©AstraZeneca 2016

The next wave of scientific innovation 23

IMED functions

Transforming clinical practice and patient outcomes in chronic respiratory diseases

Highlights We delivered – Collaboration results that significantly contributed to several projects in our portfolio (PI3K, MALT1 andRORg, as well as LTC4S (with our partner Orexo) – Three major new collaborations – 12 publications from collaborations We delivered – Recruitment of a Chief Scientist with worldleading scientific record (Gary Anderson) – 10 out of 12 new recruits directly from academia with strong science background – 71 publications in major peer-reviewed journals We delivered – Solid portfolio progress in all phases of development (from target selection to end of Phase II) – Two projects with positive Phase II data out of which one was chosen for internal progression into Phase III (PT010 Triple COPD)

Transformation timeline

Today 2015+ Win in Inhaled

The future 2020+ Transform disease management

Lead with innovative precision This year, we started to research a firstin-class, inhaled on-demand treatment to prevent exacerbations. As a result, the inhaled immunomodulator AZD9412 (recombinant IFN-β1a) progressed to Phase II evaluation in severe asthmatics. In-licensed from Synairgen, AZD9412 is being developed as an inhaled, on-demand therapy for patients with a history of exacerbations following respiratory viral infection symptoms. Such a targeted treatment approach could provide a major breakthrough in preventing morbidity in asthma, and potentially COPD patients.

People spotlight – a new generation of RIA iMed scientists

Our first rising star is Madelene Lindqvist, who joins us after four years as a postdoctoral fellow at Harvard Medical School, bringing with her profound expertise in adaptive immune mechanisms. Madelene has worked on T follicular helper cells and their role in context of HIV infection, which resulted in several publications in high-impact journals including Science Translational Medicine, Journal of Clinical Investigation, Nature Medicine and Nature Communications.

The recruitment of Kumar Krishnaswamy from Yale University School of Medicine even further strengthens the immunomodulation research in RIA. At Yale, Kumar’s postdoctoral research over the last four years was on innate-adaptive immune crosstalk with a particular focus on the role of dendritic cell subsets driving respiratory diseases like asthma. This generated key papers published in journals such as Proceedings of the National Academy of Science of the United States of America.

A major aspect of drug discovery and development is to impact on clinical outcome through predictive science. In RIA iMed DMPK, key approaches to prediction are mathematical modelling and Quantitative Systems Biology. In this context, Hoda Sharifian joins us from ETH Zürich (Swiss Federal Institute of Technology in Zurich) and brings a unique skill set to mathematically describe key biological processes against an overlay of relevant drug pharmacokinetic data. Her PhD at ETH modelled feedback regulations in the HOG MAPK pathway based on single cell measurements, resulting in high-quality publications in Molecular Systems Biology and Integrative Biology.

The next wave of scientific innovation 25

An environment where science thrives

24 ©AstraZeneca 2016

Tomorrow 2017+ Lead with innovative “Precision” approaches

Following last year’s groundbreaking respiratory deal with Almirall, in 2015 we have advanced two dual-muscarinic antagonist/β2 agonist bronchodilators (MABAs, AZD8999 and AZD8871) into Phase Ib evaluation in asthma and COPD patients, with an intent to select the best combination partner for SGRM in COPD. These MABAs are in addition to AZD2115 and others arising from our long-standing collaboration with Pulmagen Therapeutics.

2015 also saw pre-clinical development of RIA’s first small-molecule in development in a niche indication with AZD5634 – an inhaled epithelium sodium channel blocker (iENaC).

Collaborating for science innovation

– Two progressions into Phase II (Inhaled SGRM and Inhaled IFNβ) the first of which will enable future inhaled combination treatments

2016 will see RIA’s continued scientific innovation to address significant disease severity and unmet medical need in respiratory disease beyond asthma and COPD.

Focused on the enhancement of inhaled therapeutics, stage one of RIA’s research strategy saw a number of successful developments in 2015.

IMED functions

We set out to progress the RIA project portfolio.

August saw the progression of an inhaled, dry powder formulation of AZD7594 into Phase II in asthma patients. The inhaled non-steroidal glucocorticoid receptor modulator (iSGRM) potentially provides a once-daily platform for novel antiinflammatory combinations that may induce disease modification in obstructive airway diseases. A coformulation of iSGRM with abediterol has also advanced as a ‘best-inclass’ once daily future maintenance treatment for asthma patients.

This paves the way for clinical evaluation focused on restoring airway hydration, mucociliary clearance (MCC) and improving lung function in cystic fibrosis (CF), an ultimately lethal respiratory condition resulting from the genetic dysfunction of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). In pre-clinical evaluation, AZD5634 increases airway surface liquid (ASL) in vitro, improves MCC in vivo, and thus, has the potential to rehydrate the airways, restore primary innate defence and impact upon disease progression in CF patients.

Therapy area progress

We set out to strengthen our scientific leadership in the Respiratory, Inflammation and Autoimmunity area.

Progress the portfolio Focused on the enhancement of inhaled therapeutics, stage one of RIA’s research strategy saw a number of successful developments in 2015. In June, the advancement of PT010 (a MDI-inhaled, combination of budesonide, glycopyrronium and formoterol) into Phase III clinical trials for chronic obstructive pulmonary diseases (COPD) marked a major step forward for the portfolio. This was made possible through our unique technology that creates stable co‑suspensions of drug crystals in HFA propellants using lipid-based porous particles.

Transform disease management A key project in 2015 focused on modifying the underlying pathology of disease is the inhaled toll-like receptor 9 (TLR9) agonist project in asthma. AZD1419 is an immune-stimulatory oligonucleotide analogue identified through collaboration with Dynavax Technologies in California. Pre-clinical studies have shown that stimulation of TLR9 in mice induces a long-standing protection against allergic inflammation in the lung. In healthy volunteers, inhaled AZD1419 was safe and able to stimulate the local production of Type 1 interferons in the lung, the first step in the modulation of the immune response. AZD1419 generates an interferon signal in human lung at doses lower than those causing influenzalike symptoms.

Introduction

We set out to ensure impact from new and existing collaborations.

Transforming clinical practice and patient outcomes in chronic respiratory diseases

Key RIA iMed publications in 2015

RIA iMed small-molecule pipeline end of 2015

Phase IIa

Phase IIb

Phase III

AZD7594/Abediterol SGRM/LABA

AZD1419 Inhaled TLR9

AZD9412 Inhaled IFN-beta

Abediterol LABA

PT010 Triple MDI (COPD)

AZD7594/tbd SGRM/MABA

AZD7986 DPP1

AZD7594 Inhaled SGRM

PT010 Triple MDI (Asthma)

PT001 LAMA

AZD5634 iENaC (CF)

AZD8871 MABA

AZD7624 Inhaled P38

RDEA3170 URAT1 (Gout)

PT003 LABA/LAMA

AZD0284 RORg (LN)

AZD8999 MABA

Lesinurad URAT1 (Gout)

AZD9567 Oral SGRM

Seys L, Verhamme F, Schinwald A, Hammad H, Cunoosamy D, Bantsimba-Malanda C, Sabirsh A, McCall E, Flavell L, Herbst R, Provoost S, Lambrecht B, Joos G, Brusselle G, Bracke K

Clinical Pharmacology and Therapeutics

Physiologically based pharmacokinetic modelling in drug discovery and development. A pharmaceutical industry perspective

Jones HM, Chen Y, Gibson C, Heimbach T, Parrott N.J, Peters SA, Snoeys J, Upreti VV, Zheng M, Hall SD

Journal of Controlled Release

Specific accumulation of orally administered redox nanotherapeutics in the inflamed colon reducing inflammation with doseresponse efficacy

Vong LB, Mo J, Abrahamsson B, Yukio N

Mucosal Immunology

The Axl receptor tyrosine kinase is a discriminator of macrophage function in the  inflamed lung

Fujimori T, Grabiec AM, Kaur M, Bell TJ, Fujino N, Cook PC, Svedberg FR, MacDonald AS, Maciewicz RA, Singh D, Hussell, T

Structure

Ligand binding mechanism in steroid receptors: from conserved plasticity to differential evolutionary constraints

Köhler C

Journal of Cheminformatics

Target prediction utilising negative bioactivity data covering large chemical space

Mervin LH, Afzal AM, Drakakis G, Lewis R, Engkvist O, Bender A

Molecular Pharmaceutics

Fast and general method to predict the physicochemical properties of drug-like molecules using the integral equation theory of molecular liquids

Palmer DS, Misin M, Fedorov MV, Llinas A

Journal of Leukocyte Biology

Targeting neutrophilic inflammation in severe neutrophilic asthma: can we target the disease-relevant neutrophil phenotype

Bruijnzeel PL, Uddin M, Koenderman L

American Journal of Respiratory Cell and Molecular Biology

Temporal and spatial expression of transforming growth factor-beta following progressive exposure to tobacco smoke in spontaneously hypertensive rats

Hoang L, Bolton S, Wang L, Kenyon N, Nguyen Y, Smiley-Jewell S, Pinkerton K

Journal of Pharmacology and Experimental Therapeutics

A novel in vivo receptor occupancy methodology for the glucocorticoid receptor: toward an improved understanding of lung pharmacokinetic/pharmacodynamic relationships

Boger E, Ewing P, Eriksson UG, Fihn BM, Chappell M, Evans N, Fridén M

Disease Area Asthma

RA

COPD

Other

Key RIA iMed collaborations in 2015 Introduced in 2015

Ongoing with 2015 achieved milestones

University of Southampton, UK

State Key Lab of Respiratory Disease, Guangzhou Medical College, China

Catholic University of Leuven, Belgium

Access to a large number of clinically well-characterized patients to help map somatic mutations in COPD patients.

Aims to identify trigger factors for COPD exacerbations as well as exacerbation phenotypes/endotype. Provides ‘real world’ COPD cohort and information on true exacerbations rates future clinical trials.

Determine in vitro and in vivo role of MALT1’s protease activity and scaffolding role in immune cell’s function. Key high-impact papers published in 2015.

University of Helsinki, Finland

Uppsala University, Sweden

GLAZGo Discovery Centre, Glasgow, UK

Insights into how RORg inhibitors modulate the function and plasticity of Th17 cells isolated from both patients with autoimmune diseases and healthy individuals.

Development of a lung slice methodology to study mechanisms of inhaled drug retention in the lung and allow tailoring of inhalation pharmacokinetics to provide long-lasting duration of effect. Contributing fundamental knowledge and leadership in the field of inhalation science.

The RIA iMed’s collaboration with Glasgow University’s Institute of Infection, Immunity and Inflammation is breaking new ground in this area. This collaborative enterprise has been fully functional for the last 18 months, and has already yielded 15 joint projects.

26 ©AstraZeneca 2016

The next wave of scientific innovation 27

An environment where science thrives

Role of B cell activating factor (BAFF) in chronic obstructive pulmonary disease

Collaborating for science innovation

American Journal of Respiratory and Critical Care Medicine

IMED functions

Authors

Therapy area progress

Phase I

Title

Introduction

Pre-clinical Dev

Publication

Key RIA iMed publications in 2015

Nicholls DJ, Wiley K, Dainty I, MacIntosh F, Phillips C, Gaw A, Mårdh CK

British Journal of Clinical Pharmacolology

The effect of a selective CXCR2 antagonist (AZD5069) on human blood neutrophil count and innate immune functions

Jurcevic S, Humfrey C, Uddin M, Warrington S, Larsson B, Keen C

Drug Metabolism and Disposition

Lipid peroxide mediated oxidative rearrangement of the pyrazinone carboxamide core of neutrophil elastase inhibitor AZD9819 in blood plasma samples

Gu C, Lewis RJ, Wells AS, Svensson PH, Hosagrahara VP, Johnsson E, Hallström G

Respiratory Medicine

Neutrophil extracellular trap formation and extracellular DNA in sputum of stable COPD patients

Pedersen F, Marwitz S, Holz O, Kirsten A, Bahmer T, Waschki B, Magnussen H, Rabe KF, Goldmann T, Uddin M, Watz H

ChemMedChem

Benzoxazepines achieve potent suppression of IL-17 release in human T-Helper 17 (TH17) cells through an induced-fit binding mode to the nuclear receptor RORγ

Olsson RI, Xue Y, von Berg S, Aagaard A, McPheat J, Hansson EL, Bernström J, Hansson P, Jirholt J, Grindebacke H, Leffler A, Chen R, Xiong Y, Ge H, Hansson TG, Narjes F

Journal of Pharmaceutical Sciences

Development of a novel lung slice methodology for profiling of inhaled compounds

Bäckström E, Lundqvist A, Boger E, Svanberg P, Ewing P,Hammarlund-Udenaes M,Fridén M

Bioorganic and Medicinal Chemical Letters

Discovery and evaluation of a novel monocyclic series of CXCR2 antagonists

Austin RP, Bennion C, Bonnert RV, Cheema L, Cook AR, Cox RJ, Ebden MR, Gaw A, Grime K, Meghani P, Nicholls D, Phillips C, Smith N, Steele J, Stonehouse JP

Expert Review of Respiratory Medicine

Immunology, genetics and microbiota in the COPD pathophysiology: potential scope for patient stratification

Malhotra R, Olsson H

Journal of Thoracic Disease

Study on risk factors and phenotypes of acute exacerbations of COPD in Guangzhou, China – design and baseline characteristics

Zhou Y, Bruijnzeel PLB, McCrae C, Zheng J, Nihlen U, Zhou R, Van Geest M, Nilsson A, Hadzovic S, Huhn M, Taib Z, Gu Y, Xie J, Ran P, Chen R, Zhong N

Science Translational Medicine

Antibodies to influenza nucleoprotein crossreact with human hypocretin receptor 2

Ahmed SS, Volkmuth W, Duca J, Corti L, Pallaoro M, Pezzicoli A, Karle A, Rigat F, Rappuoli R, Narasimhan V, Julkunen I, Vuorela A, Vaarala O, Nohynek H, Pasini FL, Montomoli E, Trombetta C, Adams CM, Rothbard J, Steinman L

Science

Infectious disease. Life-threatening influenza and impaired interferon amplification in human IRF7 deficiency

Ciancanelli MJ, Huang SX, Luthra P, Garner H, Itan Y, Volpi S, Lafaille FG, Trouillet C, Schmolke M, Albrecht RA, Israelsson E, Lim HK, Casadio M, Hermesh T, Lorenzo L, Leung LW, Pedergnana V, Boisson B, Okada S, Picard C, Ringuier B, Troussier F, et al

28 ©AstraZeneca 2016

An environment where science thrives

Pharmacological characterisation of AZD5069, a slowly reversible CXC chemokine receptor 2 antagonist

Collaborating for science innovation

Journal of Pharmacology and Experimental Therapeutics

IMED functions

Authors

Therapy area progress

Title

Introduction

Publication

The next wave of scientific innovation 29

Case study

A major innovation for respiratory medicine research PT010 speeds into Phase III

The secret behind the co‑suspension technology

Global burden, personal struggle

The Pearl within

The idea behind the co-suspension technology used in PT010 employs tiny porous floating particles, to which the crystals associate. With the floating particles holding their stability, there is no interaction between the medicines, which remain in a uniform suspension long after the simple inversion of the inhaler, meaning a consistent and correct dosage is inhaled every time. With the co-suspension technology, all strengths and combinations of a drug delivers the same aerosol performance. This attribute is very important to regulatory agencies and is a requirement for interpreting whether pivotal clinical data have met the 'combination rule', which requires that the combination product be superior to its components.

There is a major global and individual unmet medical need in COPD. The World Health Organisation predicts the fast-growing lung disease to become the third leading cause of death by 2030, and it can severely affect quality of life for those in its grip.

In June 2013, AstraZeneca completed the acquisition of Pearl Therapeutics, which is focused on the development of inhaled small-molecule therapeutics for respiratory disease. Pearl brought to the AstraZeneca family the innovative co‑suspension technology used in PT010.

When symptoms worsen, patients become so short of breath they’re unable to undertake day-to-day tasks; struggling to climb the stairs, do the laundry, or participate in family life. The fast disease progression and often late diagnosis means that many sufferers find current treatments inadequate – failing to open the airways sufficiently to relieve the distressing effects of COPD.

Case study

August 2015 brought a positive Phase III investment decision for PT010; AstraZeneca’s fixed dosed triple for the treatment of COPD using Pearl’s innovative co-suspension technology. If successful, this will offer an improvement for patients suffering from a debilitating disease where true advances have been few and far between. PT010 combines three wellestablished compounds – a LABA (formoterol), LAMA (glycopyrronium) and an inhaled corticosteroid (ICS) (budesonide) in a totally new way that aims to enhance patient adherence and improve clinical outcomes.

The inhaler challenge In respiratory disease, inhalers are crucial to ensure delivery of the drug into the lung tissue, but persistent problems limit their therapeutic potential. To ensure consistency and interpretation of clinical outcomes, the products must have consistent in-vitro delivered dose and aerosolization properties, such as particle size distribution of the active drug. Historically, achieving this consistency has been particularly challenging for both dry powder inhalers (DPI) as well as with conventional formulations of pressurized metered-dose inhalers (pMDI) that combine multiple drugs. The co-suspension crystal technology for pMDIs was developed to address exactly this challenge.

From dual to triple in record time Patients with moderate or severe COPD commonly need two or three different medicines to effectively relieve their breathlessness. Adherence is key, and Pearl’s co-suspension technology seems to offer a solution to the delivery challenge for fixed-dose combinations. The first step was to test the technology for PT003, a dual-combination pMDI of the two bronchodilators LABA and LAMA. This successful clinical development programme led to an NDA in June 2015 and the results paved the way for taking the technology one step further – to develop a fixed dose triple for COPD patients containing a LABA, a LAMA and a corticosteroid. PT010 combines – in a single inhaler – crystals of two long-acting bronchodilators, LABA and LAMA with the ICS budesonide for immediate relief of symptoms. Relying on the extensive evidence for the three monotherapies coupled with the positive data on PT003, the clinical team were able to design a Phase III programme that was able to select and confirm the doses for the individual components, enhancing it with dose-ranging studies that included budesonide. The Phase II studies were executed quickly due to the flexible approach of the teams, and by relying on the porous particle technology that allowed for such speed. “As a physician it’s great to see how innovative technology can breathe new life into long-established medicines for the benefit of patients. As a scientist, I’m envisioning a future treatment paradigm where we marry this technology with novel pathways and compounds which tackles different aspects of the underlying cause of the disease. To me, that’s the essence of ‘What science can do’.” Maarten Kraan, VP RIA iMed “The journey so far with Pearl and the cosuspension technology has been a tremendous experience. We have successfully combined the know-how and muscle of the big, with the speed, agility and courageousness of the small. I’m really excited to bring the results to patients and physicians” Martin Olovsson, VP Inhaled Respiratory, GPPS

“There is a huge unmet medical need in COPD. The beauty of the cosuspension technology is that we’ve been able to do what others have tried and failed to do – to combine three drugs in one inhaler while retaining their efficacy. I believe that with this technology, AstraZeneca once again proves their ambition to lead the way in respiratory medicine, and others will have to follow”  olin Reisner, Head of Respiratory GMed & Chief C Medical Officer, Pearl Therapeutics

30 ©AstraZeneca 2016

The next wave of scientific innovation 31

Cardiovascular & Metabolic Diseases iMed AstraZeneca’s strategy in CVMD focuses on ways to reduce morbidity, mortality and organ damage by addressing multiple risk factors across cardiovascular (CV) disease, diabetes and chronic kidney disease indications. The patient-centric approach is reinforced by science-led lifecycle management programmes and technologies, including early research into regenerative methods.

R

32 ©AstraZeneca 2016

The next wave of scientific innovation 33

Cardiovascular & Metabolic Diseases iMed

Marcus Schindler, VP CVMD iMed

Opposite Cardiac-regenerating muscle cells

Amongst this year’s achievements is the continued improvement of the quality and breadth of our key collaborations that provides us with

Therapy area progress

Top Marcus Schindler, VP CVMD iMed

the opportunity to expand into novel scientific areas whilst maintaining a clear scientific focus. As an example; together with Regulus Therapeutics our CVMD scientists successfully progressed the novel anti-microRNA compound AZD4076 for the treatment of non-alcoholic steatohepatitis (NASH) into first-time-in-man (FTIM). At the same time, our strategic collaboration with Ionis Pharmaceuticals provided us with the complementary toolbox of antisense oligonucleotides to address targets unsuitable for smallmolecule drug discovery in CVMD. Our continued collaboration with Moderna Therapeutics showed promising results of the effect of VEGF-A modRNA in cardiac regeneration.

Introduction

“2015 was a year of achievements for CVMD iMed. We delivered exciting progress in our pipeline, continued to strengthen our new modalities platform and enhanced our capabilities with new collaborations with world-leading academic institutions and biotech companies. Our work is beginning to show the therapeutic potential of regenerative approaches in cardiovascular and metabolic diseases.”

Throughout 2015, CVMD iMed continued to advance and break new ground with new and established projects, creating the next wave of scientific innovation. By making use of our extensive knowledge and expertise in the discovery and development of small molecules we have further expanded into a ‘new modalities’ platform of modified mRNA, micro RNA, and antisense oligonucleotides. CVMD iMed is now pursuing drug targets using all of these modalities, small molecules and combinations thereof.

IMED functions

We established 13 new collaborations that gave us the opportunity to work with new techniques, and that enhanced our capabilities and expertise. Our collaborations make a significant impact on our R&D pipeline in CVMD and we are looking forward to sharing and advancing our findings in 2016.

We set out to enhance our scientific reputation and demonstrate scientific leadership.

We delivered 93 new publications in major peer-reviewed journals of which 50 in high-quality journals and three in the world’s leading journals with particularly high-impact factor. Two of our collaborations (with Shanghai Institutes for Biological Sciences and with University of Michigan) each resulted in high-impact publications strengthening our scientific reputation. Our impact on the scientific community could also be seen by the degree of citations of our publications as well as attention from social media. Significant results reflecting the impressive advancement of our discovery and clinical stage programmes were presented at key conferences in 2015 including European Society of Cardiology (ESC), European Association for the Study of Diabetes (EASD), American Diabetes Association (ADA), American Chemical Society (ACS) and American Society of Nephrology (ASN).

We set out to strengthen our diabetes research and pipeline.

We initiated an exciting five-year collaboration with Harvard Stem Cell Institute to bring in their novel breakthrough technique which creates human insulin producing β-cells from stem cells. In record time our CVMD scientists successfully set up and mastered this technique, which is an immense achievement. The cells will be used in screens of AstraZeneca’s compound library and in the search for new treatments for diabetes. The collaboration also aims to better understand how the function of β-cells declines in diabetes and research findings will be made available to the broader scientific community through peer-reviewed publications.

The next wave of scientific innovation 35

An environment where science thrives

34 ©AstraZeneca 2016

We set out to expand our scientific leadership in CVMD by collaborating with the best science outside AstraZeneca.

Collaborating for science innovation

Highlights

Taking the road less travelled by making a difference with regenerative medicine and RNA therapeutics

2015 was an exciting year and we are looking forward to further developing our science in 2016. Every member of CVMD iMed can be proud of our leaps forward.

An environment where science thrives

36 ©AstraZeneca 2016

Diabetes/NASH Non-alcoholic steatohepatitis (NASH) is inflammation and damage to the liver caused by a build up of fat in the liver. It is part of a group of diseases known as non-alcoholic fatty liver diseases. Some people with NASH have no symptoms while in others, the fat build-up causes inflammation, cell damage and in some cases cirrhosis, to a point where the liver cannot work properly. It is not known exactly what causes NASH, but it is thought be caused by any number of factors, including environmental, lifestyle and genetics. NASH risk factors include obesity, insulin resistance (type 2 diabetes) high cholesterol, high triglycerides, and metabolic syndrome. Currently, there are no approved drugs for NASH despite the imminent progress to liver failure of these patients if left untreated. Our scientists are aiming to bridge this unmet medical need with the new modalities drug AZD4076.

Dr Magnone’s expertise is a crucial part of the research for CVMD iMed. With the ongoing paradigm shift from ‘blockbuster’ therapies towards a more personalised form of medicine, based on sophisticated patient stratification where prescribed drugs are based on specific biological phenotypes rather than clinical characteristics, her expertise is right on target. Chiara has led several programmes aimed at identifying patients’ molecular phenotypes predictive of disease progression and in response to investigational treatments as well as pre-clinical programmes for the development of novel personalised healthcare drugs. In addition, her experience from the fields of insulin resistance, obesity, type 2 diabetes, NASH and chronic kidney disease truly underlines the value she brings to CVMD iMed.

Collaborating for science innovation

In parallel, the team is using cardiac stem cells for screening of drug candidates; the aim is to identify targets and pathways involved in inducing and augmenting the human body’s ability to regenerate and repair damaged cardiac tissues. From these screens small-molecule programmes were identified and are now being validated using human cells and animal models of heart failure. The broad range of modalities including small and large molecules, antisense technology, CRISPR and modified mRNA available to our scientists enables investigation and targeting new potential and previously undrugable targets.

Top Dividing pancreatic beta cells

To further explore the possibilities of identifying new treatments for diabetes a collaboration with the Harvard Stem Cell Institute was initiated. In diabetes, pancreatic β-cells are destroyed by an autoimmune response (type 1) or the β-cells either fail to function properly or their numbers decrease (type 2). In the search for diabetes treatments, human β-cells are a great asset; however, these cells are extremely limited in number and availability. With this collaboration led by HSCI co‑chairman and Howard Hughes Medical Institute Investigator, Professor Doug Melton, our CVMD scientists have access to a technique which allows potentially limitless quantities of β-cells to be produced from humaninduced pluripotent stem cells generated directly from adult cells. These cells would be similar in all important aspects to those found in healthy individuals and can be utilised for a multitude of research purposes.

Maria Chiara Magnone joined AstraZeneca in 2014 as Head of Translational Sciences, CVMD iMed where she is responsible for delivering human target validation, biomarkers and personalized healthcare (PHC) hypothesis generation across the CVMD portfolio. Chiara brings over 12 years’ experience in the area of CVMD biomarkers, personalized healthcare and target translation and was recruited to CVMD iMed from her position as Translational Medicine/Biomarker Lead at Roche and has previously held positions at Serono research and Novartis Pharma.

IMED functions

In collaboration with Moderna Therapeutics, our scientists have access to modified mRNA (modRNA), which is an attractive modality for the investigation of the local production of paracrine factors known to be important for stem cell activation and differentiation. If successful, this approach has the potential to stimulate the generation of new cardiac tissue and reverse disease in patients with heart failure. One of the lead modRNA projects, VEGF-A has shown promising results in animal models. A single injection of VEGF-A modRNA significantly increased vascular density, reduced scar area and improved the cardiac function in mice. In addition, VEGF-A stimulated cardiac stem cells (EPDCs) and induced a fate switch in these cells towards endothelial cells and to some extent to cardiomyocytes. These data demonstrate the regenerative potential of VEGF-A modRNA in the heart. The VEGF-A project is now moving to first-time-in-man to assess safety and tolerability as well as pharmacokinetics and local blood flow response, a key biological mechanism of VEGF-A.

Our pre-clinical studies have shown that inhibition of miR-103/107 by AZD4076 in mouse models, dramatically decreases liver triglyceride content and improves insulin sensitivity in both liver and peripheral tissues. AZD4076 has, therefore, the potential to act as an efficacious insulin sensitising therapy for NASH in patients with type 2 diabetes. With the collaborative effort of CVMD scientists and our partner Regulus Therapeutics Inc, we have just initiated dosing in a first-time-in-man Phase I clinical study. If successful, our compound would be a first-in-class treatment for NASH.

People spotlight

Therapy area progress

Heart failure In the cardiovascular arena CVMD scientists are targeting the growing challenge of cardiac dysfunction and heart failure. In heart failure the heart’s capacity deteriorate over time and there are currently no treatment options to reverse or even address its severe and progressive nature. A key feature of heart failure is damage due to the gradually increasing loss of cardiac tissue such as cardiomyocytes and blood vessels and the aim of the CVMD research in this area is to find a way to help the damaged heart to repair itself. It was recently shown that progenitor cells, which may play a role in cardiac repair, are present in the adult heart. Our focus is to identify targets and pathways involved in the activation and differentiation of these progenitor cells.

Right Maria Chiara Magnone

Introduction

or function, has been shown to be significantly altered or dysregulated in many disease states, including oncology, fibrosis, metabolic diseases, and immune-inflammatory diseases. Targeting microRNAs with anti-miRs – chemically modified single-stranded oligonucleotides such as AZD4076 – offers a unique approach to treating disease by modulating entire biological pathways and may become a new and major class of drugs with broad therapeutic application.

AZD4076 is a GalNAc-conjugated antimiR-103/107 oligonucleotide which was originally discovered in an alliance with Regulus Therapeutics. MicroRNAs (miR) are small RNA molecules, typically 20 to 25 nucleotides in length that do not encode proteins but rather regulate gene expression. More than 800 miRs have been identified in the human genome, and over two-thirds of all human genes are believed to be regulated by miRs. MiR expression,

The next wave of scientific innovation 37

CVMD iMed small and large-molecule pipeline end of 2015

Key CVMD iMed publications in 2015

Phase I

Phase II

AZD4831 (MPO) HFpEF

MEDI8111 (Rh Factor II) Trauma/Bleeding

AZD5718 * Cardiovascular

AZD4076 (miR103/107) * NASH

AZD4901 (hormone modulator) Polycystic Ovarian Syndrome

* Project progressed to current Phase in 2015

Key CVMD iMed collaborations in 2015 Moderna Therapeutics, Cambridge, US

Harvard Stem Cell Institute, a collaboration with Prof. Dough Melton aimed at finding regenerative drugs using inducible pluripotent stem cells and this year started differentiation protocol to utilise in industrial applications.

A strategic collaboration to discover and develop antisense oligonucleotide (ASO) therapies for cardiovascular, metabolic and renal diseases. AstraZeneca and Ionis also continue their collaboration to discover new targeted-delivery approaches to access more disease-relevant tissues for oligonucleotide therapeutics. The new CVMD collaboration further supports AstraZeneca’s strategic approach in these therapeutic areas in developing novel RNA-targeted treatments.

An alliance initiated in 2013 to discover and develop mRNA therapeutics for the treatment of cardiovascular, metabolic and renal diseases as well as cancer. The first project in the alliance, a VEGF-A encoding modRNA, is on track to enter into clinical phase in 2016 as a potential therapy for patients with cardiovascular disease.

INSERM, Paris, France

The Alliance established in 2012 to discover and develop microRNA therapeutics in CVMD and Oncology has delivered a candidate drug, AZD4076. AZD4076 is a GalNAc conjugated anti-miRNA 103/107 oligonucleotide being developed for treatment of NASH in diabetic patients.

The aim of the collaboration is to advance understanding of type 2 diabetes and chronic kidney disease (CKD) and develop new treatments based on this knowledge. The first collaboration with Professor Frédéric Jassier will aim at better understanding the complexities of mineral corticoid receptor activity as a potential treatment for CKD. The second collaboration with Professor Dominique Langin will explore pharmacological ways to prevent adipose tissue release of lipid into the circulation, to normalize fat deposition and increase insulin sensitivity in peripheral tissues. And thirdly, we will collaborate with Dr Raphaël Scharfmann to develop models of human β-cells which have lost their ability to produce and release insulin, to better understand the biology of this effect and how it can be corrected through treatment.

38 ©AstraZeneca 2016

Li Q, Yang R, Huang X, Zhang H, He L, Nie Y, Hu S, Yan Y, Wang QD, Lui KO, Zhou B

Blood

Structural and functional characterisation of a specific antidote for ticagrelor

Buchanan A, Newton P, Pehrsson S, Inghardt T, Antonsson T, Svensson P, Sjögren T, Öster L, Janefeldt A, Sandinge AS, Keyes F, Austin M, Spooner J, Gennemark P, Penney M, Howells G, Vaughan T, Nylander S

Development

How to make a cardiomyocyte

Spaeter D, Hansson EM, Zang L, Chien KR

Stem Cells Translational Medicine

Human iPSC-derived cardiac progenitor cells in phenotypic screening: a transforming growth factor-B type 1 receptor kinase inhibitor induces efficient cardiac differentiation

Drowley L, Koonce C, Peel S, Jonebring A, Plowright AT, Kattman SJ, Andersson H, Anson B, Swanson BJ, Wang QD, Brolen G

Nature Medicine

c-kit+ cells adopt vascular endothelial but not epithelial cell fates during lung maintenance and repair

Liu Q, Huang X, Zhang H, Tian X, He L, Yang R, Yan Y, Wang QD, Gillich A, Zhou B.

Cell Metabolism

SerpinB1 promotes pancreatic B cell proliferation

El Ouaamari A, Dirice E, Gedeon N, Hu J, Zhou JY, Shirakawa J, Hou L, Goodman J, Karampelias C, Qiang G, Boucher J, Martinez R, Gritsenko MA, De Jesus DF, Kahraman S, Bhatt S, Smith RD, Beer HD, Jungtrakoon P, Gong Y, Goldfine AB, Liew CW, Doria A, Andersson O, Qian WJ, Remold-O’Donnell E, Kulkarni RN

Science Translational Medicine

Tissue transcriptome-driven identification of epidermal growth factor as a chronic kidney disease biomarker

Ju W, Nair V, Smith S, Zhu L, Shedden K, Song PXK, Mariani LH, Eichinger FH, Berthier CC, Randolph A, Yi-Chun Lai J, Zhou Y, Hawkins JJ, Bitzer M, Sampson MG, Thier M, Solier C, Duran-Pacheco GC, Duchateau-Nguyen G, Essioux L, Schott B, Formentini I, Magnone MC, Bobadilla M, Cohen CD, Bagnasco SM, Barisoni L, Lv J, Zhang H, Wang HY, Brosius FC, Gadegbeku CA, Kretzler M

Angewandte Chemie

Scalable synthesis of piperazines enabled by visible-light irradiation and aluminium organometallics

Suárez-Pantiga S, Colas K, Johansson MJ, Mendoza A

Kidney International

Inhibition of the purinergic P2X7 receptor improves renal perfusion in angiotensin-IIinfused rats

Menzies RI, Howarth AR, Unwin RJ, Tam FW, Mullins JJ, Bailey MA

Structure

Ligand binding mechanism in steroid receptors; from conserved plasticity to differential evolutionary constraints

Edman K, Hogner A, Hussein A, Bjursell M, Aagaard A, Bäckström S, Wissler L, Jellesmark-Jensen T, Cavallin A, Karlsson U, Nilsson E, Lecina D, Takahashi R, Grebner C, Lepistö M, Guallar V

Journal of Medicinal Chemistry

Discovery of AZD6642, an inhibitor of 5-lipoxygenase activating protein (FLAP) for the treatment of inflammatory diseases

Lemurell M, Ulander J, Winiwarter S, Dahlén A, Davidsson Ö, Emtenäs H, Broddefalk J, Swanson M, Hovdal D, Plowright A, Pettersen A, Landergren M, Barlind J, Llinas A, Herslöf M, Drmota T, Sigfridsson K, Moses S, Whatling C

Diabetes Care

Contemporary risk estimates of three HbA1c variables for myocardial infarction in 101,799 patients following diagnosis of type 2 diabetes

Olsson M, Schnecke V, Cabrera C, Skrtic S, Lind M

Drug Discovery Today

Targeting the podocyte to treat kidney disease

Lal M, Young K, Andag U

Arteriosclerosis, Thrombosis, and Vascular Biology

Ticagrelor protects the heart against reperfusion injury and improves remodelling after myocardial infarction

Ye Y, Birnbaum GD, Perez-Polo JR, Nanhwan MK, Nylander S, Birnbaum Y

University of Michigan, US

A collaboration with Professor Matthias Kretzler in the area of chronic kidney disease (CKD). The collaboration will tackle the challenging area of identifying novel therapeutic targets for the treatment of CKD, focusing on the use of patient tissue and validation of pre-clinical models. We will also seek translatable animal models and predictive biomarkers for patient segmentation in-clinic, through a world-leading source (covering >2500 patients and seven animal models). Deliveries: human target validation data package for all portfolio projects and pre-TSID project, supporting decisions on project progression and animal models selection.

The next wave of scientific innovation 39

An environment where science thrives

Regulus Therapeutics, San Diego, US

Genetic lineage tracing identifies in situ kit‑expressing cardiomyocytes

Collaborating for science innovation

Ionis Pharmaceuticals, Carlsbad, US

Cell Research

IMED functions

Harvard University, Boston, US

Authors

Therapy area progress

AZD8601 * Cardiovascular

Title

Introduction

Pre-clinical

Publication

Neuroscience iMed The Neuroscience iMed model is unique in that it is dynamic and fully externalised, forming partnerships with leading-edge academic researchers, foundations and companies to create a portfolio of discovery and early development projects in neurological disease.

40 ©AstraZeneca 2016

The next wave of scientific innovation 41

Neuroscience iMed

John Dunlop, VP Neuroscience iMed Opposite Brain scan Top John Dunlop, VP Neuroscience iMed

PEOPLE SPOTLIGHT

To date, MEDI1814 has a good safety profile and is well tolerated. No serious adverse events have been reported and there is dose-proportional serum PK, as predicted for IgG1. Most importantly, is the demonstration of selectivity for Aβ42 versus Aβ40 in CSF. The study continues into 2016 with single and multiple dose cohorts by intravenous and subcutaneous route of administration. In parallel, clinical trials of AZD3293, an oral potent small-molecule inhibitor of β secretase cleaving enzyme (BACE), have continued to progress. A co-development effort between AstraZeneca and Eli Lilly and Company (Lilly), AMARANTH is Phase II/III study of a BACE inhibitor currently in development as a potential treatment for Alzheimer’s disease. AZD3293 has been shown in Phase I studies to reduce levels of amyloid-beta in the CSF of Alzheimer’s patients. Inhibiting BACE is predicted to prevent the formation of amyloid plaque and eventually slow the progression of the disease. The studies are examining the safety and efficacy of AZD3293 compared with placebo in the treatment of early Alzheimer’s disease. The study is progressing toward a Phase III ID in 2016.

Deposition of beta amyloid (Aβ) in the brain is a pathological hallmark of Alzheimer’s disease. There are two major Aβ isoforms: the 42-residue Aβ42 and the 40-residue Aβ40. Several studies have demonstrated that the Aβ42 species is both more toxic to neurons and more aggregate prone, despite the Aβ40 species being more predominant. AstraZeneca has been developing our selective monoclonal antibody against Aβ42, MEDI1814, and our most recent data demonstrate a selective and dose-dependent suppression of CSF Abeta1-42 but not Aβ40 in Phase 1 studies in Alzheimer’s disease patients. This is a compelling demonstration of mechanism of action and potential for therapeutic differentiation.

An environment where science thrives

42 ©AstraZeneca 2016

– Dose-dependent decrease in free Aβ42 in CSF following single IV doses

Collaborating for science innovation

Mike Perkinton in our Discovery team has been a key individual in the collaboration with Eliezer Masliah at UCSD and also in driving our synuclein programme. Mike has demonstrated scientific innovation with the leading emergent hypothesis of protein spreading, or cell-to-cell transmission, as a key pathological driver of disease progression in chronic neurodegenerative diseases including Parkinson’s disease. Mike’s work in providing the critical validation data supporting this hypothesis for our synuclein-targeted agent, which resulted in a key portfolio transition for this programme in 2015.

AstraZeneca has a unique, externalised approach to neuroscience drug discovery and development, partnering to advance the most exciting scientific opportunities in areas of high unmet medical need. In November, the team presented new data on MEDI1814, a humanised monoclonal antibody (mAb) selectively targeted to Aβ42, at the Clinical Trials on Alzheimer’s Disease (CTAD) 2015 conference in Barcelona. This event brought together world leaders in Alzheimer’s disease to discuss new results, candidate therapeutics, and methodological issues important to the development of the next generation of therapies.

– Selective target engagement following single IV doses

IMED functions

Making headway in tackling Alzheimer’s disease

– Dose-dependent increase in total plasma Aβ42 following single IV doses

Therapy area progress

Bottom Receptor in the human brain

The team presented at CTAD the findings of tolerability and preliminary pharmacodynamics studies after single doses of MEDI1814 in mild-moderate Alzheimer’s disease. MEDI1814 shows: Introduction

“In 2015, we continued to embrace our entrepreneurial and externalised approach with a number of new partnerships. These are particularly in the early portfolio and use our unique model to build for sustained delivery.”

2015 has been a year where we have really seen the benefit and impact of the AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience. This unique collaboration, established just two years ago, exemplifies our model of collaboration and externalisation. In this case, it is with a local group based in Boston, just a short distance from our Cambridge (MA) office. We now see tangible impact on both our portfolio and our science. This is best illustrated by the target validation and screening hit characterisation for the KCC2 modulator programme, performed in close collaboration with our IMED Discovery Sciences colleagues. KCC2 is a critical determinant of neuronal excitability in the brain and implicated in epilepsies and Amyotrophic Lateral Sclerosis (ALS). In addition to project support, the lab has delivered high-quality publications in the Journal of Neuroscience and the Proceedings of the National Academy of Sciences characterising the basic biology of this target.

The next wave of scientific innovation 43

Neuroscience iMed clinical pipeline end of 2015

Phase I

MEDI7352 Analgesia

AZD8108 NMDA antagonist Suicidal ideation

Application of cross-species PET imaging to assess neurotransmitter release in brain

Finnema SJ, Scheinin M, Shahid M, Lehto J, Borroni E, Bang-Andersen B, Sallinen J, Wong E, Farde L, Halldin C, Grimwood S

MEDI1814 amyloid beta mAb Alzheimer’s disease

AZD3241 myeloperoxidase inhibitor Multiple System Atrophy

Brain

Effect of the myeloperoxidase inhibitor AZD3241 on microglia: a PET study in Parkinson’s disease

Jucaite A, Svennigsson P, Rinne JO, Cselényi Z, Varnäs K, Johnström P, Amini N, Kirjavainen A, Helin S, Minkwitz M, Kugler AR, Posener JA, Budd S, Halldin C, Varrone A, Farde L

Molecular Psychiatry

Misassembly of non-mutant disrupted-inschizophrenia 1 (DISC1) protein is linked to altered dopamine homeostasis and behavioral deficits

Brandon N

Molecular Psychiatry

Early postnatal GABAA receptor modulation reverses deficits in neuronal maturation in a conditional neurodevelopmental mouse model of DISC1

Saito A, Taniguchi Y, Rannals M, Merfeld E, Ballinger M, Minori K, Ohtani Y, Gurley D, Sedlak T, Cross A, Moss S, Brandon N, Maher B, Kamiya A

Neuron

BrainSeq: neurogenomics to drive novel target discovery for neuropsychiatric disorders

Schubert C, O’Donnell P, Quan J, Wendland J, Hualin S, Domenici E, Essioux L, Kam-Thong T, Didriksen M, Matsumoto M, Saito T, Brandon N, Cross A, Wang Q, Heon Shin J, Jaffe A, Jia Y, Straub R, Deep-Soboslay A, Hyde T, Kleinman J, Weinberger D

Journal of Alzheimer’s Disease

AZD3293: a novel, orally active BACE1 inhibitor with high potency and permeability and markedly slow off-rate kinetics

Eketjäll S, Janson J, Kaspersson K, Bogstedt A, Jeppsson F, Fälting J, Budd Haeberlein S, Kugler AR, Alexander RC, Cebers G

Nature Neuroscience

The cellular targets of antidepressants

Brandon N, McKay R

Cellular Signalling

Uncovering the function of disrupted in schizophrenia 1 through interactions with the cAMP phosphodiesterase PDE4: contributions of the Houslay lab to molecular psychiatry

Brandon N

Biological Psychiatry

The novel metabotropic glutamate receptor 2 positive allosteric modulator, AZD8529, decreases nicotine self-administration and relapse in squirrel monkeys

Justinova Z, Panlilo L, Secci M, Redhi G, Schindler C, Cross A, Mrzljak L, Medd A, Shaham Y, Goberg S

Neuropharmacology

State-dependent alterations in sleep/wake architecture and antipsychoticlike activity by the M4 Positive Allosteric Modulator VU0467154

Gould R, Nedelcovych M, Gong X, Tsai E, Bridges T, Wood M, Bubser M, Daniels J, Duggan M, Brandon N, Dunlop J, Wood M, Ivarsson M, Noetzel M, Niswender C, Lindsley C, Conn P, Jones C

British Journal of Pharmacology

Quetiapine and its metabolite norquetiapine translation from in vitro pharmacology to in vivo efficacy in rodent models

Cross A, Widzowski D, Maciag C, Zacco A, Hudzik T, Liu J, Nyberg S, Wood M

Journal of Clinical Psychopharmacology

AZD6280, a novel partial GABA-A receptor modulator, demonstrates a pharmacodynamically selective effect profile in healthy male volunteers

Chen X, Jacobs G, de Kam M, Jaeger J, Lappalainen J, Maruff P, Smith M, Cross A, Cohen A, van Gerven J

Proceedings of the National Academy of Sciences of the United States of America

KCC2 activity is critical in limiting the onset and severity of status epilepticus

Silayeva L, Deeb T, Hines R, Kelley M, Munoz M, Lee H, Brandon N, Dunlop J, Maguire J, Davies P, Moss S

The Journal of Neuroscience

Selective inhibition of KCC2 leads to hyperexcitability and epileptiform discharges in hippocampal slices and in vivo.

Sivakumaran S, Cardarelli RA, Maguire J, Kelley MR, Silayeva L, Morrow DH, Mukherjee 2, Moore YE, Mather RJ, Duggan ME, Brandon NJ, Dunlop J, Zicha S, Moss SJ, Deeb TZ

MEDI1341 Parkinson’s disease AZD3293* beta-secretase inhibitor Alzheimer’s disease *Partnered project

Key Neuroscience iMed collaborations in 2015 University of Pennsylvania and Stanford University, US

Eolas Therapeutics & NIH, Carlsbad, US

Trinity College Dublin, Ireland

Partnership funded by Target-ALS to discover new targets modifying the toxicity of ALS causative and risk genes.

Three-way partnership with biotech company Eolas and the NIH to advance orexin1 receptor antagonists in the area of addiction disorders, funded by the NIH Blueprint Neurotherapeutics network.

Generation of new mechanistic data on human Aβ and physiological measures of cognition.

Imperial College London, UK

University of Sussex, UK

Eli Lilly, Indianapolis, US

Preclinical data characterizing novel bifunctional mAb in analgesia.

Working with the university’s Translational Drug Discovery Group to explore novel GABA receptor modulators in Huntington’s disease and anxiety disorders, funded by the MRC and Wellcome Trust.

Phase II/III trial of AZD3293, an oral potent small-molecule inhibitor of BACE – good progress towards Phase III ID in 2016.

44 ©AstraZeneca 2016

The next wave of scientific innovation 45

An environment where science thrives

Psychopharmacology

Phase II

Collaborating for science innovation

Authors

IMED functions

Title

Therapy area progress

Journal/publication

Introduction

Pre-clinical

Key Neuroscience iMed publications in 2015

Discovery Sciences Applying world-leading expertise to drive the identification of quality targets, hits, leads and drug candidates that will be safe and efficacious in the clinic.

46 ©AstraZeneca 2016

The next wave of scientific innovation 47

Discovery Sciences

Highlights

Mike Snowden, VP Discovery Sciences

Engineered cell lines to selected IMED projects, e.g. the SIK (salt-induced kinase) project for use in evaluating which one of three isoenzymes was the drug target and secondly, provided a mutant form of EGFR (C797S) to support that project.

Conduct a phenotypic screen using a larger collection of compounds than typically used for such assays.

A phenotypic screen that used 120,000 compounds that is about ten-fold more than typically used. We executed this technically challenging screen using a seven-day proliferation assay that involved more than 50 steps using a novel beta cell, EndoC-βH1 and a high-throughput flow cytometry readout. The output from this screen will provide chemical equity for the Melton collaboration, which is seeking modulators of beta cell growth.

Enhance the compound collection at AstraZeneca by the exchange of chemical equity with peer companies.

A first-in-world exchange of 210,000 compounds between AstraZeneca and Sanofi, and a further 25,000 compounds exchanged with a leading agrochemical company.

Deliver the strategy for compound management in Cambridge Biomedical Campus.

A first-class strategy aligned with several exciting collaborations. The first stage is the relocation of our solid store from Alderley Park to a world-renown expert, located in Europe. The temperature-controlled stores that house our liquid compound collection are being developed in collaboration with Brooks. With another partner, LabCyte, AstraZeneca is working on groundbreaking technology that will deliver compounds from tubes to plates using acoustic technology.

Establish a world-class HTS capability at Cambridge Biomedical Campus.

A strategic collaboration with HiRes of Boston to deliver a modular automation solution for HTS. Such systems provide a high degree of flexibility as the units are mounted on carts that can be moved around; to be easily reconfigured to fit the screening modality required.

Provide computational biology and mathematical modelling work packages to support IMED projects.

A tailor-made image analysis software package for a novel hepatotoxicity model that used cells grown in 3D, as spheroids. The package calculated the size/volume of these cell clusters, which was a critical measurement parameter that relied completely on the experts in-house, as there was not an off-the-shelf solution. In addition, we continue to explore statistical methods that reduce the number of animals used in experiments. The use of micro-sampling that takes ‘snapshots’ of the experimental design is reducing the number of animals required.

To deliver more predictive liver toxicity assays.

A novel hepatotoxicity model that cultures HepG2 cells grown in 3D rather than the conventional 2D/flat dish system. Cells grown under 3D conditions form spheroids rather than a monolayer, this 3D phenotype is more liver-like with cells expressing drugmetabolising enzymes (cytochrome p450s) unlike those grown in 2D conditions. The assay has been validated with known liver toxins and compares very favourably against the gold standard assay primary human liver cells. The new assay is about three times faster than the outsourced human liver assay and ten-fold cheaper.

Collaborating for science innovation

Investigate the use of precise genome editing (CRISPR/Cas9) to deliver genetically engineered cell lines to IMED and external collaborators.

IMED functions

 

To complement our Hit Identification platforms, and to ensure that we reserve them for the most validated drug discovery projects, we pushed ahead at pace with our ambitious plans to revolutionise our pre-clinical target validation capabilities through the widespread use of precise genome editing in both cellular and in pre-clinical animal model systems. Through collaboration with the very best external scientists in the world, and through significant internal investment we delivered an unprecedented number of both cellular and animal models to AstraZeneca projects and, importantly, developed the technological platform to optimise gene knockout generation, resulting in two patent filings and the potential for a number of high-impact publications in 2016. 2015 was a bumper year for publication for Discovery Sciences, with over 120 peerreviewed papers, including over 50 high-quality and seven high-impact publications.

We delivered

Therapy area progress

2015 has been an incredible year for Discovery Sciences. We continued with the development of our small-molecule Hit Identification capabilities and employed plate-based high-throughput screening (HTS), DNA encoded libraries and fragment-based screening to more of our internal projects than ever before. We made our HTS compound collection available to a record number of academic collaborators, and initiated exciting projects with Brooks, Labcyte and HiRes to bring state-of-the-art robotic compound management and screening platforms to AstraZeneca as we prepare for the move of our HTS capability to the Cambridge Biomedical Campus, and to our partners in the Centre for Lead Discovery, Cancer Research UK and the Medical Research Council.

We set out to

Introduction

“Discovery Sciences goes from strength to strength with another year of exceptional delivery and innovation. The Hit Identification platforms we developed in 2014 continue to provide world-class support for IMED small-molecule discovery, and we augmented this capability with the development of one of the most advanced genome editing-based target validation platforms in the industry. An incredible year indeed!”

An environment where science thrives

Left Mike Snowden, VP Discovery Sciences

48 ©AstraZeneca 2016

The next wave of scientific innovation 49

Key Discovery Sciences collaborations in 2015

People spotlight

Use of cryoEM to determine the structure of large protein complexes. AstraZeneca is part of ten-partner consortium.

Collaboration to deliver tube-to-plate compound dispensing using acoustic technology (Labcyte) and tailor-made plastic ware (Brooks).

Research agreement that allows AstraZeneca access to Pelago’s thermal shift technology for use in target engagement and target identification studies.

HiRes, Boston, US

Labcyte and Waters, Sunnyvale, US and Elstree, UK

Knut and Alice Wallenberg Foundation and Royal Institute of Technology, Stockholm, Sweden

Collaboration to design and deliver modular, flexible HTS automation solutions for the Lead Discovery Centre at CBC.

Collaboration to use acoustic liquid dispensing technology (Labcyte) to add samples to a mass spectrometer (Waters) for use in biochemical and cellular assays.

Explore new targets for disease research through the area of secretomics.

Key Discovery Sciences publications in 2015 Authors

Nature Communications

Responding to the challenge of untreatable gonorrhea: ETX0914, a first-in-class agent with a distinct mechanism-of-action against bacterial Type II topoisomerases

Basarab GS, Kern GH, McNulty J, Mueller JP, Lawrence K, Vishwanathan K, Alm RA, Doig P, Gardner H, Gowravaram M, Huband M, Kutschke A, Lahiri SD, Tommasi R, Newman J, Schuck V, Singh R, Kimzey A, Morningstar M, Barvian K

Nature Communications

Structural and dynamic insights into the energetics of activation loop rearrangement in FGFR1 kinase

Klein T, Vajpai N, Phillips J, Davies G, Holdgate G, Phillips C, Tucker J, Norman R, Scott A, Higazi D, Lowe D, Breeze A

Nature Communications

Oxidation of the alarmin IL-33 regulates ST2dependent inflammation

E. Suzanne Cohen, Ian C. Scott, Jayesh B. Majithiya, Laura Rapley, Benjamin P. Kemp, Elizabeth England, D. Gareth Rees, Catherine L. Overed-Sayer, Joanne Woods, Nicholas J. Bond, Christel Séguy Veyssier, Kevin J. Embrey, Dorothy A. Sims, Michael R. Snaith, Katherine A. Vousden, Martin D. Strain, Denice T. Y. Chan, Sara Carmen, Catherine E. Huntington, Liz Flavell, Jianqing Xu, Bojana Popovic, Christopher E. Brightling, Tristan J. Vaughan, Robin Butler, David C. Lowe, Daniel R. Higazi, Dominic J. Corkill, Richard D. May, Matthew A. Sleeman,* and Tomas Mustelin*

Nature Reviews Drug Discovery

An analysis of the attrition of drug candidates from four major pharmaceutical companies

Michael J. Waring, John Arrowsmith, Andrew R. Leach, Paul D. Leeson, Sam Mandrell, Robert M. Owen, Garry Pairaudeau, William D. Pennie, Stephen D. Pickett, Jibo Wang, Owen Wallace, Alex Weir

Nature Reviews Drug Discovery

Towards a hit for every target

Rees S, Janzen W, Gribbon P, Pairaudeau G, Birmingham K

Science Advances

Structural basis of Lewisb antigen binding by the Helicobacter pylori adhesin BabA

Hage N, Howard T, Phillips C, Brassington C, Overman R, Debreczeni J, Gellert P, Stolnik G, Winkler G, Falcone F

The next wave of scientific innovation 51

An environment where science thrives

Title

Collaborating for science innovation

Publication

IMED functions

Jon Wingfield joined AstraZeneca in 2000 as part of the oncology function, tasked with the delivery of automation to support secondary screening activities. After building delivery-focused screening teams, he moved into Discovery Sciences upon its formation in 2010. As a Principal Scientist, he is responsible for the strategic delivery of objectives and ensuring the projects have high visibility with both internal and external scientific communities. Jon uses his experience of drug discovery to showcase the potential value of this revolutionary technology platform with collaborative partners.

Pelago, Stockholm, Sweden

Therapy area progress

50 ©AstraZeneca 2016

Martin Bachman is a postdoc within Discovery Sciences, joining AstraZeneca in April 2015 after completing his DPhil at Cambridge University. Martin’s main area of research, prior to AstraZeneca, was in the role of DNA modifications in epigenetics. He already has published in Nature Chemistry and Proceedings of the National Academy of Sciences, showing the use of mass spectrometry as a tool to test biological hypotheses. Martin supports the delivery of data for potential scientific applications and is currently building assays to demonstrate the system’s potential as a high-throughput screening platform.

Labcyte and Brooks, Cambridge, UK

Introduction

Ian Sinclair joined AstraZeneca from Waters in 1999 to manage and develop the growth of Open Access LCMS across Alderley Park. Since 2007, he has examined the use of charged aerosol detection (CAD) for use in compound management quality assurance. More recently, he has studied strategies to measure and improve quality within large compound collections. Ian brings an invaluable technical depth of knowledge while managing the project and liaison with our technical counterparts at Labcyte and Waters.

Laboratory of Molecular Biology, Cambridge, UK

Case study

Behind the scenes

Acoustic mass spectrometer Our technique of choice for high-resolution microscopy

The technology supporting our science

The stats

– High throughput: 10,000 data points generated per hour, with three samples per second going into the acoustic mass spectrometer versus one sample every ten seconds with a standard mass spectrometer platform

– Allows us to generate data points at 10,000 data points per hour

– Small sample volumes required: only 2µl samples needed, meaning it is possible to get multiple samples more easily – No cross-contamination: Using acoustics to move samples means no cross-contamination risk as nothing touches any surfaces

–T  he acoustics fire at 500Hz frequency (500 times per second) – that means we fire in 500 bursts of droplets every second – For some analytes, we only need ~160 droplet bursts from a sample to generate enough ions – this means we can deliver samples to the acoustic mass spectrometer at a rate of three per second. Currently, the best highthroughput screening platform only sends one sample every ten seconds to the acoustic mass spectrometer

Case study

The facts

Mass spectrometry (or mass spec) is an analytical chemistry technique that helps identify the amount and type of chemicals present in a sample by measuring the mass-to-charge ratio and abundance of gas-phase ions. As standard, mass specs have three components – a component to convert sample from liquid to gaseous form. Then a component to ionise the sample (hit with high energy to turn to +ve or –ve ions), then the component to detect quantities of each ion (peaks of each ion seen). Instead of using a needle to aspirate and spray the samples as with standard mass specs, the acoustic mass spectrometer sends a sonic pulse through liquid creating a ‘mountain of liquid on the surface’. For compound handling, a 2.5 nanoLitre droplet is generated and fired from the liquid surface. For acoustic mass spectrometer we send a second pulse through the mountain of liquid and this explodes the 2.5nL droplet into hundreds of femtoLitre droplets. In effect, sending a tornado of droplets through a charged field to generate a stream of ionised particles into the acoustic mass spectrometer.

The scientist perspective “When you see the droplets flying and you see the science behind it – it’s astonishingly cool. It’s revolutionary. It allows us to work at very high throughput and with very small sample volumes – we only need 2µl of sample. Using this screening system we can screen more compounds or more targets for the same cost. For some assays we could reduce the cost of high-throughput screening by 80%. AstraZeneca has brought together a world leading supplier of mass spec technology (Waters) and the global leader in acoustic droplet ejection technology (Labcyte) to deliver this revolutionary platform. It required the vision of AstraZeneca’s scientific leaders to recognise the potential of this system. We took the idea to the partners, suggesting that it is possible to use acoustics in a different way in mass spec. Being open about the work we are doing has shown the wider scientific community that AstraZeneca is prepared to invest in groundbreaking science, we are not just making a small change in mass spec, but we are leading a potential revolution in mass spec screening. As with any novel area of research this project carries an element of risk. Again, our openness shows that AstraZeneca is prepared to take scientific risks. We’re engaging the broader scientific community early in the process, and delivering real benefit to the community overall. The acoustic mass spectrometer platform won the 2015 Innovation Award from the Society for Laboratory Automation and Screening. This is awarded to the podium presentation at the annual conference that shows the most innovation and impact in screening. AstraZeneca is the first non-US-based company to receive this award. The webcast of the podium presentation was made available to members after the meeting, and the society received so many requests to access this presentation that they made it free to non-members for a period, which is unprecedented. Subsequently, the society has decided to make the presentation available for free to the science community for two years. We have also had a paper about the platform accepted in a peerreviewed journal (Journal of Laboratory Automation).” Jonathan Wingfield, Principal Scientist, Discovery Sciences

Opposite Acoustic mass spectrometer

1

1nL is 1 millionth of a millilitre (or 1 trillionth of a litre). One femtolitre is 1 millionth of a nanolitre (or 0.000000000000001 litre)

52 ©AstraZeneca 2016

Above Jonathan Wingfield, Principal Scientist Discovery Sciences

The next wave of scientific innovation 53

Drug Safety & Metabolism Driving our science to bring better, safer medicines to patients sooner.

54 ©AstraZeneca 2016

The next wave of scientific innovation 55

Drug Safety & Metabolism Introduction

“2015 was an exceptional year for DSM. On top of delivering against all project timelines in the IMED and Global Medicines Development portfolio, I’ve been amazed with the progress of our teams, who have made particularly great progress with oligonucleotide-based therapies this year where we are already seeing really encouraging results.” Stefan Platz, VP Global Drug Safety & Metabolism

Opposite DNA Holliday junction molecular model

56 ©AstraZeneca 2016

We continue to build our network within the local Cambridge community, and as well as hosting and sponsoring a number of local seminars and conferences, in 2015 we formed a pre-competitive alliance with Cambridge University and GlaxoSmithKline on medicines safety. In addition to supporting the complete IMED pipeline, and through a time of significant footprint change, 2015 was an exceptional year for DSM; here are some of our key highlights:

– Enabling Technologies Next Generation Sequencing and health patch pilots initiated, a human tissue lead appointed and increased bioinformatics support – People and Organisation Introduction of a PCRA engagement programme for DSM employees to help embed our PCRA strategy – CKD Pilot – CKD is the newest core disease area within CVMD and the pilot offers the opportunity to build a patient-centric safety foundation to identify key areas of concerns and map the potential need of new capabilities – COPD Pilot – The COPD pilot offers an exciting opportunity to develop a more proactive approach to assessing risk pre-clinically – Internal and External Influence The launch and implementation of pilot studies in two key disease areas in collaboration with the wider IMED organisation – Data Information and Integration The agreement to recruit a Lead Information Officer, as well as extensive analysis of data capture and flow of information

The next wave of scientific innovation 57

An environment where science thrives

We also initiated several key academic collaborations to strengthen our scientific reputation in the area of safety science, including one with the MRC Toxicology Unit (“Investigations into the interference of nucleotide modalities and their delivery systems on the translational machinery, nucleotide stress and immune signalling pathways”), and another with Uppsala University on expanding molecular imaging technologies to improve drug safety and efficacy understanding.

The global DSM team also worked to define, build and lead the Patient Centric Risk Assessment (PCRA) strategy. 35 DSM colleagues, approximately 10% of the department, in four PCRA workstreams aimed to bring to life a common purpose, the patient, across DSM.

The four Patient Centric Risk Assessment workstreams and key 2015 outcomes

Collaborating for science innovation

Collaborative working was a focus for DSM in 2015. Within the ‘New Modalities’ space, a key strategic area for DSM, we worked with external partners to establish deeper understanding of oligonucleotide and modRNA therapeutics.

In Cambridge, our Laboratory Animal Sciences team, who provide invaluable support across IMED, became part of the new Research Support Facility (RSF), a combined IMED and MedImmune team, which is setting the direction for future collaborative working between the two groups. Our team also helped transition the Medical Research Council Laboratory of Molecular Biology to their new base in Cambridge.

IMED functions

Drug Safety and Metabolism (DSM) is an IMED function contributing to the entire value chain. Structured into six functions and with a purpose to drive science to bring better, safer medicines to patients sooner, we support and enable pipeline progression in AstraZeneca’s core therapeutic areas. With our vast range of expertise we deliver in silico, in vitro and in vivo data to deliver target and chemical risk assessments as well as metabolic quantitative translational safety models, including imaging for functional and pathological safety signals. We design and deliver tailored safety pharmacology and toxicology packages to support project decisionmaking and enable safe progression through clinical development.

Therapy area progress

Top Stefan Platz, VP Globa Drug Safety & Metabolism

Highlights We delivered

Consider the safety implications of CRISPR as a therapeutic modality and the use of CRISPR technologies for safety model build.

Collaborations with world-leading scientists for animal model builds, off-target analysis, DNA repair and targeted delivery. Our scientists have presented both nationally and internationally on the safety concerns associated with CRISPR and we have set up a crosscompany workgroup with key leaders to consider CRISPR safety.

Become the industry leaders in the application of modelling and simulation (M&S) to address drug safety.

A strong translational M&S team with a demonstrated track record of project impact, particularly in oncology. In addition, we have begun to construct novel safety modelling platforms in-house and through collaborations. Finally, we have increased our level of visibility and leadership among industry peers by, for example, chairing the platform session at the American Conference of Pharmacometrics (premier society meeting) on systems modelling for drug safety.

Support the progression of the first AstraZeneca modRNA therapeutic to candidate drug stage.

Progression with AZD8601 to candidate drug stage in October of 2015, and with the drug entering GLP toxicology studies in Q1 2016, we will enable first-in-human trials later in 2016.

Progress the first anti-microRNA oligonucleotide drug into man.

Safety data and documentation that enabled the first human dose to be administered in Phase I less than nine months from candidate drug identification.

– Flexible – can be encoded to produce any protein – Adaptable – proteins can act intracellularly or be secreted – Functional – diverse and potent therapeutic potential

58 ©AstraZeneca 2016

From his experience in this US biotech environment, Patrik learnt the importance of sharing challenges through continuous and open debate. Acknowledging team efforts, working together, celebrating achievements and daring to try new things were prevalent traits in this professional environment. Equally applicable to scientific research and the Californian surf culture, Patrik realised that you need to lose your preconceptions and engage with the experts – what may at first appear simple may in fact have a more complex reality. Since his return to Sweden, Patrik has contributed to the strategic Ionis collaborations in the CVMD and targeted delivery areas and with new ideas for the Moderna platform.

The next wave of scientific innovation 59

An environment where science thrives

The new modelling approach that has been developed generates accurate predictions for both intravenous and subcutaneous dosing and the biodistribution of LNPs. Unlike classical pharmacokinetic models, these mechanistic PBPK models are capable of describing the biodistribution of LNPs. Their ability to predict tissue/targetspecific concentration profiles provides a model-based framework that can be used to maximise therapeutic index. Of particular significance is the fact that their physiological basis allows for more sophisticated approaches for model translation from one species to another.

As the potential delivery method of choice for oligo therapies, exosomes are critical for the successful development of CRISPR, enabling efficient transfer of Cas9 and guide-RNA. However, endogenous exosomal protein and RNA contamination raises safety concerns. Led by Mick Fellows, the DSM Discovery Safety department has set up a new collaboration in 2015 with leaders in the field to research and develop technologies that enable clean exosome preparation and to assess the risk from contamination whilst maintaining delivery efficiency. This is expected to significantly contribute to the translation of exosomes from the lab to the clinic and will place AstraZeneca in a unique position to lead in new modality delivery systems for the next generation of therapeutics.

Collaborating for science innovation

DSM have been investigating the physiologically-based pharmacokinetic (PBPK) modelling approach for predicting biodistribution of mRNA-loaded lipid nanoparticles (LNPs) from a safety testing perspective. PBPK models are particularly well-suited for supporting model-translation from one scenario to another, e.g. species, exposure route, dose. This is because LNP kinetics differ from that of small molecules and are typically very non-linear and impacted distinctly through interactions with different tissues and cells.

In August 2014, Patrik Andersson moved his Gothenburg family of five to the biotech cluster in San Diego for a 12-month secondment to learn more about oligonucleotide therapeutics. This drug platform has different properties to small molecules and they target RNA rather than proteins, leading to different challenges, approaches and screening cascades. Patrik’s objectives were to support ongoing projects in CVMD and Oncology and identify new collaboration opportunities for DSM.

IMED functions

Successful development and delivery of these models as well as securing a key collaboration with a German biotech company, TissUse, to explore the next wave of scientific innovation, which will connect individual organ units together.

CRISPR (clustered regularly interspaced short palindromic repeats) is a genomeediting tool that allows fast and precise changes to be made in specific genes. The technology has two components – a homing device to a specific section of DNA (guide-RNA) and enzymatic ‘scissors’ that cut DNA (Cas9 nuclease). In the cell nucleus, the guide-RNA sequence directs the Cas9 nuclease to cause double-stranded breaks in the target DNA sequence. By harnessing the cell’s own DNA-repair apparatus, the gene being targeted can be altered either by deleting it, adding nucleotides to it or by turning its activity on or off.

People spotlight

Therapy area progress

Create rat and dog liver-on-a-chip models as part of our ongoing organson-a-chip development.

AstraZeneca has been partnering with Moderna Therapeutics since 2013 to discover, develop and commercialise pioneering mRNA Therapeutics™. This unique approach uses proprietary mRNA containing nucleotide analogues, which are designed to stimulate the body’s natural ability to produce intracellular and secreted therapeutic proteins without triggering an innate immune response. Modified mRNA may dramatically reduce the time and expense associated with creating therapeutic proteins using current recombinant technologies. Moreover, as a therapeutic device, modified mRNA are:

Another line of investigation that has begun in 2015 is the use of exosome delivery systems to explore potential safety concerns. Exosomes are endogenous nanocarriers of RNA and proteins that mediate communication between cells. As many cancers are characterised by exosome-associated alterations, exosomes are considered to be relevant drug delivery vehicles for cancer treatment.

Introduction

We set out to

Thinking differently about safety using CRISPR and mRNA modelling

Key Drug Safety & Metabolism collaborations in 2015 University of Cambridge, UK

University of Cambridge, UK

Strengthening the IMED futures workstream, the TissUse collaboration has been established to explore the value of human bone marrow-on-achip model to predict drug-induced hematotoxicity and genotoxicity.

Andreas Bender in Chemistry. The project will explore bioinformatics algorithms for predicting drug-drug interactions based on clinical adverse event database mining.

Experimental Medicine Ph.D. student. This proposal was selected as the first clinical PhD post of the ‘Experimental Medicine’ initiative and is on the subject of quantitative systems models of human cardiovascular physiology.

Ludwig Maximillians University, Munich, Germany

Uppsala University, Sweden

This collaboration aims to develop and validate a new fast-responding and non-invasive CYP3A biomarker in urine, 1B hydroxydeoxycholic acid, for CYP3A inhibition DDI studies.

This collaboration will look to generate pig models with inducible expression of Cas9 and with transgenic expression of humanised PCSK9.

This collaboration is expanding molecular imaging technologies to improve drug safety and efficacy understanding and to discover and develop pathology biomarkers.

University of Cambridge, UK

AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3, with early evidence of clinical activity in lymphoma and lung cancer

Hong D, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, Fowler N, Zhou T, Schmidt J, Jo M, Lee SJ, Yamashita M, Hughes SG, Fayad L, Piha-Paul S, Nadella MVP, Mohseni M, Lawson D, Reimer C, Blakey DC, Xiao X, Hsu J, Revenko A, Monia BP, MacLeod AR

Nature Communications

Triaminopyrimidine is a fast-killing and longacting antimalarial clinical candidate

Hameed S, Solapure S, Patil V, Henrich P, Magistrado P, Bharath S, Murugan K, Viswanath P, Puttur J, Srivastava A, Bellale E, Panduga V, Shanbag G, Awasthy D, Landge S, et al

British Journal of Pharmacology

Effects of acute and chronic sunitinib treatment on cardiac function and calcium/ calmodulin dependent protein kinase II

Mooney L, Skinner M, Coker SJ, Currie S

Molecular Pharmaceutics

Flagging drugs that inhibit the bile salt export pump

Montanari F, Pinto M, Khunweeraphong N, Wlcek K, Sohail MI, Noeske T, Boyer S, Chiba P, Stieger B, Kuchler K, Ecker GF

British Journal of Pharmacology

Future technology insight: mass spectrometry imaging as a tool in drug development

Cobice D, Goodwin R, Andren P, Nilsson A, Mackay C, Andrew R

Archives of Toxicology

Hepatic effects of repeated oral administration of diclofenac to hepatic cytochrome P-450 reductase null (HRN?) and wild type mice

Akingbasote J, Foster A, Wilson I, Sarda S, Jones H, Gerry Kenna J

Analytical Chemistry

Mapping drug distribution in brain tissue using liquid extraction surface analysis mass spectrometry imaging

Swales J, Tucker J, Spreadborough M, Iverson S, Clench M, Webborn P, Goodwin R

CPT: Pharmacometrics & Systems Pharmacology

Modeling and simulation approaches for cardiovascular function and their role in safety assessment

Collins TA, Bergenholm L, Abdulla T, Yates JWT, Evans N, Chappell MJ, Mettetal JT

Toxicological Sciences

Re-evaluation of the mutagenic response to phosphorothioate nucleotides in human lymphoblastoid TK6 cells

Saleh A, Priestley C, Gooderham N, Fellows M

Toxicological Sciences

Correlation of in vivo versus in vitro benchmark doses (BMDs) derived from micronucleus test data: a proof of concept study

Soeteman-Hernández L, Fellows M, Johnson G, Slob W

Chemical Research in Toxicology

Aortic binding of AZD5248, mechanistic insight and reactivity assays to support lead optimzation

Bragg R, Brocklehurst S, Gustafsson F, Goodman J, Hickling K, MacFaul P, Swallow S, Tugwood J

The next wave of scientific innovation 61

An environment where science thrives

Science Translational Medicine

Collaborating for science innovation

60 ©AstraZeneca 2016

Authors

IMED functions

This collaboration will study genetic variability in myocyte calcium handling and the consequences for drug-induced toxicity.

Title

Therapy area progress

Karolinska Institute, Stockholm, Sweden

Publication

Introduction

TissUse, Berlin, Germany

Key Drug Safety & Metabolism publications in 2015

Personalised Healthcare and Biomarkers Personalised Healthcare is an integral part of our approach to discovering and developing new medicines; we use diagnostics and biomarkers to target our treatments to patients most likely to benefit.

62 ©AstraZeneca 2016

The next wave of scientific innovation 63

Personalised Healthcare and Biomarkers “We see Personalised Healthcare as the future of medicine – it shows us what science can do.” Ruth March, VP PHB

Opposite DNA sequence

To stay at the forefront of Personalised Healthcare science, we are constantly looking for novel diagnostic technologies that can help us deliver better solutions to patients. For example, the EGFR

Highlights We delivered

Deliver PHC to patients: enable launch of AstraZeneca drug products linked to diagnostic tests.

Seven diagnostics launched with our diagnostic partners linked to four AstraZeneca products: – Myriad’s tumour BRCA analysis (EU) for olaparib – Qiagen’s EGFR companion diagnostic test (US), and circulating tumour DNA EGFR plasma test (EU) for gefitinib

IMED functions

We set out to

Therapy area progress

Top Ruth March, VP PHB

In 2015, we have launched seven diagnostic tests linked to our products (see highlights) and led companion diagnostic development for AstraZeneca’s three FDA-approved PHC drugs.

mutation test launched in the EU for osimertinib (AZD9291) is the world’s first diagnostic test intended for both circulating tumour DNA (ctDNA) derived from plasma and tumour DNA derived from solid tissue. Indeed, we are now able to include diagnostic testing based on plasma for many drug projects, achieving a label update in China for gefitinib in 2015. Such use of plasma for testing enables up to 25% more patients without evaluable solid tumour samples to access the right treatment. In addition, we are continuing to explore diagnostics based on next-generation sequencing in several of our drug programmes, through partnerships with Illumina and Foundation Medicine. This year we have started to explore the potential of Droplet Digital PCR (ddPCR) in partnership with Sysmex-Inostics, a highly sensitive diagnostic technology.

Introduction

Personalised Healthcare is an integral part of our approach to discovering and developing new medicines; we use diagnostics and biomarkers to target our treatments to patients most likely to benefit. More than 80% of AZ’s clinical pipeline, and 95% of our small-molecule IMED pipeline, is following a Personalised Healthcare approach, with over 50 planned drug launches in the next eight years requiring a linked diagnostic test.

– Roche Molecular System’s EGFR tissue and plasma test (EU) – the world’s first diagnostic test for both circulating tumour DNA and tumour tissue, and EGFR companion diagnostic test (US) for osimertinib – Ventana’s PD-L1 diagnostic test (Class I in vitro diagnostic test) in US and in EU for durvalumab – first FDA regulated PD-L1 diagnostic – Exploration of novel diagnostic technologies, such as ctDNA, NGS and ddPCR, in nine drug projects for biomarkers, including FGFR, Akt, MET, TP53, KRAS and EGFR as well as gene panels – Shaping of the diagnostic landscape through industry-wide collaborative initiatives, such as the Stratified Medicines Innovation Working Group, ICH E18 Genomics Working Group and European Biopharmaceutical Enterprises

Collaborating for science innovation

Achieve leadership in PHC science.

– Advancing new diagnostics and treatments arising from the 100,000 Genomes project through the Genomics England public-private GENE consortium

Bring the benefits of PHC to all core therapy areas.

We are now making significant progress in extending the benefits of PHC to patients in all therapy areas: – In inflammatory disease, we are developing a handheld uric acid diagnostic test for patients with inflammatory disease – In cardiovascular and metabolic disease, we are collaborating with the Montreal Heart Institute to search the genomes of up to 80,000 patients for genes associated with cardiovascular and diabetes disease, their complications and treatment outcomes

Invest in strategic collaborations.

– 14 new collaboration agreements signed with diagnostic companies, increasing our investment to >$130m – Partnerships with leading academic centres, including the Karolinska Institute, Cambridge University, Montreal Heart Institute, and The Wellcome Trust Sanger Institute

64 ©AstraZeneca 2016

The next wave of scientific innovation 65

An environment where science thrives

– Over 30 new staff hired to enhance our diagnostic and biomarker expertise

Developing a companion diagnostic test for a breakthrough therapy

Osimertinib achieved one of the fastest clinical development programmes on record, taking less than three years from first-time-inman to first launch.

Introduction Therapy area progress

Non-small-cell lung cancer (NSCLC) is the most common form of cancer worldwide. Patients that have activating mutations of the Epidermal Growth Factor Receptor (EGFR) gene are more likely to benefit from EGFR Tyrosine Kinase Inhibitors (TKIs) such as gefitinib, and diagnostic tests for these mutations are used to select patients for first-line treatment. Although EGFR-TKIs are effective in these patients, the majority develop resistance after 10-12 months, and in around 60% of these cases, resistance is associated with the emergence of another EGFR mutation, known as T790M. Knowledge of the biological role of these mutations helped AstraZeneca to design osimertinib (AZD9291), an irreversible EGFR-TKI that targets both the sensitising mutations of EGFR and the T790M mutation that confers resistance.

Osimertinib achieved one of the fastest clinical development programmes on record, taking less than three years from first-time-in-man to first launch. One of the contributing factors of AZD9291’s success was its ability to use a companion diagnostic test to select the right patients for treatment from the earliest stages of clinical development. AstraZeneca’s PHB function partnered with Roche Molecular Systems (RMS) to develop the cobas® EGFR Mutation Test as a companion diagnostic test to identify the right patients for treatment, often accelerating delivery of essential diagnostic modules to enable fast-tracking of drug development timelines. In November 2015, osimertinib was approved by the US FDA for the treatment of patients with metastatic epidermal growth factor receptor (EGFR) T790M mutation positive non-small-cell lung cancer (NSCLC), as detected by an FDA-approved test, who have progressed on or after EGFR TKI therapy, with the companion diagnostic being approved on the same day. Just five weeks later, the Committee for Medicinal Products for Human Use of the European Medicines Agency announced a positive opinion for the same drug.

IMED functions

People spotlight

Craig Barker brings expertise to PHB from Leica in Tissue Diagnostics, including regulatory interactions and commercial markets. As the new Head of Tissue Diagnostics and member of PHB Labs leadership team based in Cambridge, he has built a team of dedicated diagnostic scientists who deliver companion diagnostic assays that select patients most likely to benefit from linked AstraZeneca drugs. Since joining PHB in January 2015, Craig has rapidly established his group’s ability to manage global deployment of the diagnostic assays and work with diagnostic partners to submit regulatory packages against tight timelines.

An environment where science thrives

66 ©AstraZeneca 2016

Carolina Haefliger is the new Head of Companion Diagnostics for Cardiovascular and Metabolic Diseases (CVMD), and is also responsible for Opportunistic therapy areas such as Neuroscience. An MD in clinical genetics with expertise in both CVMD and oncology translational science and the leadership of observational biomarker studies, Carolina joined PHB from Novartis in July 2015, and focuses on providing diagnostic options to our CVMD portfolio as well as championing strategic collaborations such as our genomics initiative with Montreal Heart Institute on behalf of the CVMD Therapy Area Leadership team. A member of PHB’s leadership team based in Gothenburg, Carolina also serves as PHB’s Medical Adviser.

Collaborating for science innovation

Joachim Reischl brings expertise in global biomarker strategy and development from Bayer, combined with scientific leadership in pharmacology and genomics. As PHB site head in Gothenburg and member of PHB’s leadership team since July 2015, Joachim heads up the new, global Policy, Portfolio and Externalisation (PPE) group that drives organisational efficiency within PHB and provides diagnostic project management across a growing portfolio. The PPE group is also responsible for providing content for AstraZeneca’s policy initiatives on Personalised Healthcare.

The next wave of scientific innovation 67

PHC adoption across AstraZeneca pipeline end of 2015

Phase II 25 New Molecular Entities

Phase III 10 New Molecular Entities

Applications under review 5 New Molecular Entities

Small molecule

Large molecule

Small molecule

Large molecule

Small molecule

Large molecule

AZD1419# TLR9 asthma

MEDI4920 CD40L-Tn3 pSS

AZD7594 Inhaled SGRM asthma

AZD9412#    Inhaled βIFN asthma/COPD

PT010 LAB/LAMA/ICS COPD

anifrolumab# TULIP IFNaR SLE

AZD7986 DPP1 COPD

MEDI5872# B7RP1 SLE



abediterol (AZD0548) LABA asthma/COPD

mavrilimumab# GM-CSFR rheumatoid arthritis

roxadustat# HIFPH anaemia CKD/ESRD

Benralizumab# IL-5R severe asthma

lesinurad URAT-1 gout

AZD8999 MABA asthma/COPD

MEDI7836 IL-13 asthma



AZD7624 Inhaled p38 inhibitor COPD

MEDI2070# IL-23 Crohns

selumetinib# SELECT-1 MEK 2L KRAS+ NSCLC

brodalumab# IL-17R psoriasis

AZD9291 AURA, AURA 2 EGFR T790M NSCLC>2L

AZD9977 MCR diabetic kidney disease

MEDI0382 GLP-1/gluagon diabetes/obesity

RDEA3170 URAT-1 hyperuricemia/gout

tralokinumab IL-13 severe asthma

Cediranib ICON 6 VEGF PSR ovarian

AZD3759 or AZD9291 BLOOM   EGFR NSCLC brain mets

MEDI6012 LCAT ACS



AZD4901 PCOS

abrilumab# α4β7 Crohns/ulcerative colitis

durvalumab#ATLANTIC PD-L1 3L NSCLC

AZD5312# androgen receptor prostate

MEDI8111 Rh-Factor II trauma/bleeding



AZD1775# Wee-1 ovarian

MEDI9929# TSLP asthma/atopic dermatitis

moxetumomab# CD22 HCL

AZD6738 ATR solid tumours

MEDI0562# hOX40 solid tumours



AZD2014 mTOR 1/2 solid tumours

MEDI-551# CD19 DLBCL

tremelimumab DETERMINE CTLA-4 mesothelioma

AZD8186 PI3Kβ solid tumours

MEDI0639# DLL-4 solid tumours



AZD4547 FGFR solid tumours

MEDI-573#   IGF metastatic breast cancer

MEDI-551# CD19 neuromyelitis optica



AZD5363# AKT breast cancer

susatoxumab (MEDI4893) staph alpha toxin SSI

AZD9150# STAT3 haems & solids

MEDI3617# ANG-2 solid tumours



savolitinib# MET pRCC

MEDI7510 sF+GLA-SE RSV prevention

AZD9496 SERD ER+ breast

MEDI565# CEA BITE tumours

AZD3241 MPO Multiple System Atrophy

MEDI8897# RSV passive prophylaxis

ATM AVI# BL/BLI SBI AZD8108 NMDA suicidal ideation





AZD3293# BACE Alzheimer’s

MEDI9447 CD73 solid tumours



AZD5847 oxazolidione TB

MEDI1814 amyloidβ Alzheimer's



CXL# BLI/cephalosporin MRSA

CAZ AVI # BLI/cephalosporin SBI/clAl/cUTI



Pipeline data correct as of 30 September 2015. Includes significant fixed dose combination projects, and parallel indications that are in a separate therapeutic area # Partnered; ¶ Registrational Phase II/III study

RIA

Oncology

CVMD

Infection, Neuroscience, Gastrointestinal

Project with PHC Approach

Key PHB collaborations in 2015 Qiagen, Hilden, Germany

Ventana, Tuscon, US

Roche Molecular Systems, Basel, Switzerland

Master Collaboration Agreement focussed on non-invasive diagnostic test using Circulating Free DNA for Non-small-cell lung cancer.

Agreement focused on prostate cancer: PTEN biomarker test, gastric cancer: ATM IHC diagnostic tissue test, and PD-L1 expression Class I device and companion diagnostic test.

Master Collaboration Agreement focused on lung cancer: KRAS mutation diagnostic test (solid tumour and plasma), breast cancer: PI3K mutation test in plasma DNA, and lung cancer: T790M EGFR mutation test (solid tumour and plasma).

Myriad, Salt Lake City, US

Illumina, Cambridge, UK

Master Collaboration Agreement focused on BRCA testing in pancreatic, lung, breast and ovarian cancers.

Partnership focused on the development of an Oncogene Panel and Therapy Selection System, and the implementation of next-generation sequencing as an FDA-approved companion diagnostic.

MEDI3902 PsI/PcrV pseudomonas MEDI-550 pandemic influenza virus vaccine MEDI8852 influenza A treatment

68 ©AstraZeneca 2016

The next wave of scientific innovation 69

An environment where science thrives

MEDI6383# Ox40 FP solid tumours

  

Collaborating for science innovation

MEDI0680 PD-1 solid tumours



  

PT003 PINNACLE LABA/LAMA COPD

IMED functions

AZD8835 PI3Kα solid tumours



  

Large molecule

Therapy area progress



  

Small molecule

Introduction

Phase I 30 New Molecular Entities

Key PHB publications in 2015 Authors

European Heart Journal

Effect of genetic variations on ticagrelor plasma levels and clinical outcomes

Varenhorst C, Eriksson E, Johansson Å, Barratt BJ, Hagström E, Åkerblom A, Syvänen AC, Becker C, James SK, Katus HA, Husted S, Steg G, Siegbahn A, Voora D, Teng R, Storey RF, Allentin L

European Journal of Nuclear Medicine and Molecular Imaging

(11)C-PBR28 imaging in multiple schlerosis patients and healthy controls: test-retest reproducibility and focal visualization of active white matter areas

Park E, Gallezot JD, Delgadillo A, Liu S, Planeta B, Lin SF, O’Connor KC, Lim KI, Lee JY, Chastre A, Chen MK, Seneca N, Leppert Dl, Huang Y, Carson RE, Pelletier D

Nature: Insight

Precision medicine, AstraZeneca’s approach

March R

Journal of Nuclear Medicine

Fazio P, Svenningsson P, Forsberg A, Jönsson EG, Amini N, Nakao R, Nag S, Halldin C, Farde L

Nature Communications

Aberrant splicing of U12-type introns is the hallmark of ZRSR2 mutant myelodysplastic syndrome

Madan V, Kanojia D, Li J, Okamoto R, Sato-Otsubo A, Kohlmann A, Sanada M, Grossmann V, Sundaresan J, Shiraishi Y, Miyano S, Thol F, Ganser A, Yang H, Haferlach T, Ogawa S, Koeffler HP

A quantitative analysis of 18F-(E)-N-(3Iodoprop-2-Enyl)-2b-carbofluoroethoxy-3b(49-methyl-phenyl) nortropane binding to the dopamine transporter in Parkinson’s disease

Journal of Cerebral Blood Flow & Metabolism

Cselény Z, Farde L

Haematologica – The Hematology Journal

Refractory anemia with ring sideroblasts and marked thrombocytosis (RARS-T) cases harbor mutations in SF3B1 or other spliceosome genes accompanied by JAK2V617F and ASXL1 mutations

Jeromin S, Haferlach T, Weissmann S, Meggendorfer M, Eder C, Nadarajah N, Alpermann T, Kohlmann A, Kern W, Haferlach C, Schnittger S

Quantification of blood flow-dependent component in estimates of beta-amyloid load obtained using quasi-steady-state standardized uptake value ratio

Journal of Leukocyte Biology

Targeting neutrophilic inflammation in severe neutrophilic asthma: can we target the disease-relevant neutrophil phenotype?

Bruijnzeel PL, Uddin M, Koenderman L

Cancer Research

Exploring the biomechanical properties of brain malignancies and their pathological determinants in vivo with magnetic resonance elastography

Jamin Y, Boult JKR, Li J, Popov S, Garteiser P, Ulloa JL, Cummings C, Box G, Eccles SA, Jones C, Waterton JC, Barnber JC, Sinkus R, Robinson SP

Chemistry of Materials

Influence of the base on Pd@MIL-101NH2(Cr) as catalyst for the Suzuki-Miyaura cross-coupling reaction

Yao Q, Bermejo Gómez A, Su J, Pascanu V, Yun Y, Zheng H, Chen H, Liu L, Abdelhamid HN, Martín-Matute B, Zou X

Brain, Behavior, and Immunity

Telomere length and outcomes in ischaemic heart failure: data from the controlled rosuvastatin multiNAtional trial in heart failure (CORONA)

Haver V, Mateo Leach I, Kjekshus J, Fox JC, Wedel H, Wikstrand J, de Boer RA, van Gilst WH, McMurray JJ, van Veldhuisen DJ, van der Harst P

Cerebrospinal fluid kynurenines in multiple sclerosis; relation to disease course and neurocognitive symptoms

Aeinehband S, Brenner P, Ståhl S, Bhat M, Fidock M, Khademi M, Olsson T, Engberg G, Jokinen J, Erhardt S, Piehl F

Radiology

Zhang WJ, Cristinacce P, Bondesson E, Nordenmark L, Young SS, Liu YZ, Singh D, Naish JH, Parker GJM

Circulation: Cardiovascular Genetics

The NLRC4 inflammasome is an important regulator of interleukin-18 levels in patients with acute coronary syndromes: a genomewide association study in the PLATO trial

Johansson Å, Eriksson N, Becker RC, Storey RF, Himmelmann A, Hagström E, Varenhorst C, Axelsson T, Barratt BJ, James SK, Katus HA, Steg G, Syvänen AC, Wallentin L, Siegbahn A

MR quantitative equilibrium signal mapping: a reliable alternative to CT in the assessment of emphysema in patients with chronic obstructive pulmonary disease

Nuclear Medicine and Molecular Imaging

β-Amyloid binding in elderly subjects with declining or stable episodic memory function measured with PET and [11C] AZD2184

Mattsson P, Forsberg A, Persson J, Nyberg L, Nilsson LG, Halldin C, Farde L

Stem Cells Translational Medicine

Concise review: workshop review: understanding and assessing the risks of stem cell-based therapies

Heslop JA, Hammond TG, Santeramo I, Piella AT, Hopp I, Zhou J, Mills, Park BK

British Journal of Clinical Pharmacology

The effect of a selective CXCR2 antagonist (AZD5069) on human blood neutrophil count and innate immune functions

Jurcevic S, Humfrey C, Uddin M, Warrington S, Larsson B, Keen C

Neuropharmacology

A PET study comparing receptor occupancy by five selective cannabinoid 1 receptor antagonists in non-human primates

Hjorth S, Karlsson C, Jucaite A, Varnäs K, Wählby Hamrén U, Johnström P, Gulyás B, Donohue SR, Pike VW, Halldin C, Farde L

European Journal of Nuclear Medicine and Molecular Imaging

Test-retest reproducibility of [(11)C]PBR28 binding to TSPO in healthy control subjects

Collste K, Forsberg A, Varrone A, Amini N, Aeinehband S, Yakushev I, Halldin C, Farde L, Cervenka S

NeuroImage

Diurnal and seasonal variation of the brain serotonin system in healthy male subjects

Matheson GJ, Schain M, Almeida R, Lundberg J, Cselényi Z, Borg J, Varrone A, Farde L, Cervenka S

Molecular Psychiatry

Contribution of non-genetic factors to dopamine and serotonin receptor availability in the adult human brain

Borg J, Cervenka S, Kuja-Halkola R, Matheson GJ, Jönsson EG, Lichtenstein P, Henningsson S, Ichimiya T, Larsson H, Stenkrona P, Halldin C, Farde L

Brain: A Journal of Neurology

Effect of the myeloperoxidase inhibitor AZD3241 on microglia: a PET study in Parkinson’s disease

Jucaite A, Svenningsson P, Rinne JO, Cselényi Z, Varnäs K, Johnström NA, Kirjavainen A, Helin S, Minkwitz M, Kugler AR, Posener JA, Budd S, Halldin C, Varrone A

Radiology

T1-weighted dynamic contrast-enhanced MR imaging of the lung in asthma: semiquantitative analysis for the assessment of contrast agent kinetic characteristics

Zhang WJ, Niven RM, Young SS, Liu YZ, Parker GJM, Naish JH

Diabetes

Increasing pyruvate dehydrogenase flux as a treatment for diabetic cardiomyopathy: a combined 13C hyperpolarized magnetic resonance and echocardiography study

Le Page LM, Rider OJ, Lewis AJ, Ball V, Clarke K, Johansson E, Carr CA, Heather LC, Tyler DJ

Cardiovascular Genetics

Differential genetic effects on statin-induced changes across low-density lipoproteinrelated measures

Chu AY, Guilianini F, Barrratt BJ, Ding B, Nyberg F, Ridker PM, Chasman DI

Chemistry – A European Journal

Influence of the base on PD@MIL-101-NH2 (CR) as catalyst for the Suzuki-Miyrua crosscoupling reaction

Carson F, Pascunu V, Bermejo Gomez A, Zhang Y, PlateroPrats AE, Zou X, Martin-Matute B

Nature Science Reports

Adaptation to acetaminophen exposure elicits major changes in expression and distribution of the hepatic proteome.

Eakins R, Walsh J, Randle L, Jenkins RE, SchuppeKoistinen I, Rose C, Starkey LP, Vasieva O, Prats N, Brillant N, Auli M, Bayliss M, Webb S, Res JA, Kitteringham NR, Goldring CE, Park BK

European Journal of Heart Failure

70 ©AstraZeneca 2016

The next wave of scientific innovation 71

An environment where science thrives

Title

Collaborating for science innovation

Publication

IMED functions

Authors

Therapy area progress

Title

Introduction

Publication

Early Clinical Development Where science meets the patient, skilled transitional clinical scientists who evaluate whether our research can change lives.

72 ©AstraZeneca 2016

The next wave of scientific innovation 73

Early Clinical Development

A strategy with three principles

“In 2015, the second year of Early Clinical Development (ECD), we created significant positive change in our strategy, people, operations and culture. ECD has attracted world-class talent globally and established a Clinical Discovery Unit (CDU). Operational simplification was reflected in substantial improvements in clinical trial cycle times, underpinned by an increasingly entrepreneurial and accountable culture.”

– Design and deliver innovative clinical studies to progress the pipeline

Tony Johnson, VP Early Clinical Development Opposite Immune response to cancer

– Accelerate human target validation (HTV) across AstraZeneca core therapeutic areas

Process improvement has been another key area of focus, with the implementation of eight new initiatives to ensure simplification and improved operational efficiencies. Compared to 2014, we have reduce our cycle times in Phase I by 28% and in Phase II by 41%. These improvements have generated significant cost reduction and 36% increased efficiency. In order to achieve such improvements, ECD has progressively evolved its culture to foster agility and accountability in decision-making coupled with an entrepreneurial ‘can do’ attitude.

Progress the pipeline across the core therapeutic areas.

Enrolment of Phase I studies for oncology (e.g. c-met inhibitor, AZD6094 and AKT inhibitor, AZD5363) were completed with encouraging data thus far; multiple Phase I and Phase II starts; RIA P38 Phase II trial achieved its interim analysis; CVMD – dosed the MCR antagonist in humans and completed the MAD study.

Implement innovative study designs.

Over 20 clinical trials incorporated an adaptive design, increasing our ability to be responsive to the evolving knowledge of our product candidates. Certain respiratory studies have been initiated with novel early phase endpoints which, in some cases, halved the required number of patients and reduced the study durations, e.g. P38 MAPK inhibitor. Scientifically innovative model-based evaluation of biomarker data has also been incorporated within our adaptive study designs to achieve earlier POM, e.g. DPP1.

Increase patient-centricity.

Real-time data capture from patients (e.g. PROACT) and real-time visualisation of clinical trial data (e.g. REACT) have guided clinical trial adaptation and more efficiently determined the right dose/regimen and patient population for our novel medicines. In 2015, REACT was further improved to become web-based and incorporate safety, efficacy, PK and biomarker data, a significant industry-leading achievement.

Foster excellence in scientific leadership.

ECD produced 124 publications – 32 of which were high quality and five were high impact, a 118% improvement from 57 publications in 2014. These demonstrated world-class early clinical development science in oncology, RIA, CVMD, Quantitative Clinical Pharmacology (QCP) and Biometrics.

Systematically integrate data from multiple sources.

QCP developed renal disease models incorporating pharmacokinetics and pharmacodynamics for lesinurad and the two xanthine oxidase inhibitors, allopurinol and febuxostat, which included the key documented influence of renal filtration on clearance of uric acid key components in gout. Using model-based simulations, it was demonstrated that lesinurad exhibits a positive benefit-risk in key clinical scenarios identified by the FDA.

Be the partner of choice to external partners.

ECD scored a rating within the top three for our managed relationships with external partners.

The next wave of scientific innovation 75

An environment where science thrives

We delivered

Collaborating for science innovation

74 ©AstraZeneca 2016

We set out to

IMED functions

Highlights

Therapy area progress

Top Tony Johnson, VP Early Clinical Development

– Integrate data from multiple sources systematically to inform research and development

We have also recruited six exceptional physician scientists from top universities to push the translational science agenda in ECD.

Introduction

ECD’s new strategy reflecting our focus on ‘where science meets the patient’ was communicated in October 2015. Skilled translational clinical scientists evaluate whether our research can change lives, dependent on three fundamental principles:

In addition, we have built strategic alliances with some of the best teaching hospitals, including Cambridge, Harvard and Manchester.

Cycle Time (number of months)

41% reduction

Industry benchmark 31

2012-14

2013-15

Rolling years

2014-16

Cycle Time (number of months)

Phase I Cycle Time

25

Industry benchmark 24 28% reduction

20 15

The Experimental Medicines Initiative at the University of Cambridge is another key collaboration for the CDU. AstraZeneca funds one PhD and two academic lecturer positions for clinicians per year. The first PhD scientist has been appointed and will start work with the Drug Safety and Metabolism (DSM) team in early 2016. The aim is to expand this model to other key academic partners. Importantly, recruitment has been a major focus for ECD in 2015. We have attracted world-class talent globally across all departments and disciplines to consolidate our geographical footprint in Cambridge (UK), Boston and Gaithersburg (US) and Gothenburg (Sweden). Recruiting and retaining world-class talent is fundamental to the innovation, creativity, dedication and execution-focus that will enable ECD to become industry leaders. During 2015, we have recruited 66 positions in ECD, 70% in Cambridge (UK), 20% in Gothenburg (Sweden), 9% in Boston (US) and 1% in Gaithersburg (US). By the end of 2015, the permanent ECD headcount was 237, an increase of 12% compared to 212 at the end of 2014.

10 5 0

2012-14

2013-15

Rolling years

2014-16

The next wave of scientific innovation 77

An environment where science thrives

76 ©AstraZeneca 2016

80 70 60 50 40 30 20 10 0

The CDU is actively looking to recruit PhD clinical scientists to further expand the AstraZeneca talent pool. Two academic clinical lecturers have been seconded from the University of Cambridge to build collaborative links in CVMD and oncology respectively. Further secondments both from within and from outside AstraZeneca are being explored.

Collaborating for science innovation

Use of the portfolio power and agility of AstraZeneca creates the opportunity to search for early scientific signals from potential therapies, alone or in combination, targeting niche patient populations. Taking a modular approach makes it possible for ECD to truly follow the science, as different combinations can be added in response to evolution in each patient’s cancer. Placeholders are added to the protocol from the outset, to create the flexibility to test combinations dependent on the specific molecular cancer drivers detected. Such an approach is especially effective for using immunotherapy agents, which are often administered in combination with targeted agents. Within the Tatton study, a rolling-arm allocation was used as a specific design feature to ensure ongoing parallel recruitment so that patients could always join an appropriate trial arm in an efficient manner. The dose-finding phase of Tatton has now been completed, while the trial is entering the expansion phase under the supervision of Global Medicines Development (GMD).

Phase II Cycle Time

People are at the heart of CDU’s function – through collaboration with leading academic institutions and by recruiting and developing talented physician scientists across therapeutic areas. Professor Stephen Rennard is Chief Clinical Scientist and a respiratory physician whose key area of interest it to recruit a large cohort of chronic obstructive pulmonary disease (COPD) patients and to characterise them fully. This will identify COPD clinical subsets and allow us to target the right drugs to the right patients.

IMED functions

We deliver innovative clinical studies Innovation in fit-for-purpose clinical trial design is a key strategic pillar for ECD. In 2015, ECD initiated its first basket trial design in the AZD9291 Tatton lung cancer study, collaborating with the oncology IMED. The creativity is reflected in multiple treatment options for patients within a single trial based on the molecular driver of each patient’s cancer. In addition, the design allowed for combinations of therapies to be tested using the extensive AstraZeneca portfolio of small and large molecules while also enabling more efficient progression through trials of each monotherapy or drug combination regimen. AstraZeneca has been an early adopter of this approach from an industry perspective.

We integrate data and inform research and development In the context of improving data integration to knowledge, the second ECD strategic pillar, significant enhancements have been delivered at the patient interface using REACT. ECD’s partnership with Tessella Technology is to develop innovative new technologies that allow datadriven, scientific decisions during, rather than on completion, of a clinical trial. This augments R&D efficiency by earlier discontinuation of those drugs with a low probability of meeting their predefined safety and/or efficacy goals. REACT was developed to enable AstraZeneca researchers to view patient information from ongoing clinical trials within 24 hours of data reaching AstraZeneca. REACT tracks laboratory tests, adverse events, and can monitor biomarker and efficacy data on both population and subject-specific levels during the course of a clinical trial. In 2015, REACT has evolved to become much more user-friendly through conversion to a webbased format. In addition, the ability to incorporate safety, efficacy, biomarkers and PK in a single study, as was achieved with a key savolitinib study, allows many scientific questions to be evaluated. This enables a data-driven, appropriate adaptation of the study design during the course of a trial and is an exciting improvement of real-time data visualisation capabilities.

The Clinical Discovery Unit was set up in 2015 by Professor Tim Eisen. Its primary purpose is to expand the capacity for translational clinical science and accelerate HTV as a key contribution for ECD.

Therapy area progress

Strategic pillars

Operations Due to ECD’s disciplined coordination between operational activity, efficiency and licence to operate, our clinical operations have improved their overall efficiency by 36% since being established in 2014. Clinical Pharmacology Unit (CPU) costs have been reduced by 12%, laboratory costs by 15% and CPU protocol deviations by 27%. Most significantly, we set out to deliver 20% improvement in our Phase I and Phase II cycle times. We achieved a reduction of 28% in Phase I (16m v industry 24m) and a reduction of 41% in Phase II (44m v Industry 31m), underscoring the enabling role of ECD across IMED’s delivery programme.

People spotlight

Introduction

We accelerate human target validation ECD is focusing increasing effort on accelerating Human Target Validation (HTV). All functions of ECD are involved and will increasingly be boosted by the development of the Clinical Discovery Unit. HTV will often be achieved in collaboration with academic partners. For example, in collaboration with Lars Lund at Karolinska, the CVMD TMU demonstrated that Myeloperoxidase (MPO)-related biomarkers outperformed NT-proBNP in predicting NYHA score. This significantly adds to previous work suggesting that MPO drives endothelial dysfunction and mortality in heart failure with preserved ejection fraction. Other CVMD TMU examples include FLAP in inflammatory disease and GPR44 in dysfunction of human pancreatic beta cells. CDU, working with key ECD partners, commenced work on a study to recruit a large cohort of patients with COPD who will be very thoroughly characterised. As well as providing greater understanding of the different disease phenotypes, this will enable segmentation of the COPD population and rapid recruitment of patients to multiple molecularly targeted Phase II studies using novel portfolio agents.

Key Early Clinical Development collaborations in 2015 National Jewish Health, Denver, US

Boston University School of Medicine, US

University of Georgia, US

Sarah Cannon Research Institute, London, UK

Karolinska Institute, Stockholm, Sweden

Investigator: Professor Johann De Bono, MD, MSc, PhD, FRCP, FMedSci. Professor of Experimental Cancer Medicine, Honorary Consultant Medical Oncologist at the Royal Marsden Hospital and Institute of Cancer Research Expert at developing molecular targeted therapies for prostate cancer patients and the projects have involved evaluating AstraZeneca novel therapeutics in prostate cancer.

Investigator: Dr Howard A. Skip Burris, III, MD, FACP. President, Clinical Operations and Chief Medical Officer, Sarah Cannon Providing AstraZeneca with clinical development expertise, access to molecular profiling data and timely, cost-efficient CRO trial management for early phase novel oncology clinical trials with potential for personalised medicine approaches.

Investigator: Professor Peter Stenvinkel, MD, PhD Renal Medicine and Kerstin Brismar, MD, PhD Growth and Metabolism Collaboration set up to provide access to clinical samples and data from several large cohorts of CKD patients as well as healthy controls, which were and will continue to be used to assess HTV in multiple targets for the management of CKD, as well as some pre-TSID CKD projects.

IMED functions

Institute of Cancer Research and The Royal Marsden Hospital, London, UK

Therapy area progress

Investigator: Assistant Professor K. Melissa Hallow, PhD. Joint appointment in College of Engineering and College of Public Health, Department of Epidemiology and Biostatistics The project involves diabetes disease modelling in collaboration with DMPK CVMD and DSM and provides support to late stage and early diabetes assets including chronic kidney disease.

Introduction

Investigator: Professor Avrum Spira, MD, MSc. Division of Computational Biomedicine, Department of Medicine This project seeks to find key gene expression and protein biomarkers that identify COPD patients with high levels of p38 MAP kinase and MEK activity in order to correlate protein kinase activity with clinical outcomes in COPD and lung cancer and to identify patients who might best respond to specific p38 and MEK inhibitors.

Investigator: Associate Professor Elena Goleva, PhD. Division of Pediatric Allergy and Immunology This project examines the role of p38 MAP kinase in steroid resistant asthma, and explores the effect of AZD7624 (inhaled p38 inhibitor) in severe steroidresistant asthma.

Title

Authors

The New England Journal of Medicine

AZD9291 in EGFR inhibitor-resistant nonsmall-cell lung cancer

Jänne PA, Hsin Yang JC, Kim DW, Planchard D, Ohe Y, Ramalingam SS, Ahn MJ, Kim SW, Su WC, Horn L, Haggstrom D, Felip E, Kim JH, Frewer P, Cantarini M, Brown KH, Dickinson PA, Ghiorghiu S, Ranson M

European Heart Journal

Effect of genetic variations on ticagrelor plasma levels and clinical outcomes

Varenhorst C, Eriksson N, Johansson Å, Barratt B, Hagström E, Åkerblom A, Syvänen AC, Becker RC, James SK, Katus HA, Husted S, Steg PG, Siegbahn A, Voora D, Teng R, Storey RF, Wallentin L

The Lancet Oncology

Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, openlabel, multicentre trial

Motzer RJ, Hutson TE, Glen H, Michaelson MD, Molina A, Eisen T, Jassem J, Zolnierek J, Maroto JP, Mellado B, Melichar B, Tomasek J, Kremer A, Kim HJ, Wood K, Dutcus C, Larkin J

Nature Reviews Cancer

VHL, the story of a tumour suppressor gene

Gossage L, Eisen T, Maher ER

Gut

Galectin-3 regulates hepatic progenitor cell expansion during liver injury

Hsieh WC, Mackinnon AC, Lu WY, Jung J, Boulter L, Henderson NC, Simpson KJ, Schotanus B, Wojtacha D, Bird TG, Medine CN, Hay DC, Sethi T, Iredale JP, Forbes SJ

78 ©AstraZeneca 2016

An environment where science thrives

Publication

Collaborating for science innovation

Key Early Clinical Development publications in 2015

The next wave of scientific innovation 79

Shaping drug development in Asia

Background

A key growth area for AstraZeneca

We enhanced our external scientific reputation through highimpact publications, presentations at major international scientific conferences, and collaborations with leading research institutions. The positive results of olaparib Phase II study in gastric cancer was published in the Journal of Clinical Oncology in 2015. Our poster on AZD3759 Phase I study was selected for oral discussion at ASCO 2015. Several of our senior scientists have been invited to join leading academic institutions such as Beijing University as faculty members.

– The dose and schedule for further clinical studies has been identified through Phase II clinical studies initiated in 2015 – China IND filing was accepted by China FDA in April 2015. This is the first category 1.1 (China Innovation) filing by AstraZeneca

Scientific leadership in action to accelerate delivery of our innovative medicines to patients in China

These plans aim to help us accelerate development of our medicines in this important market by expanding our

These investments and dedicated R&D capabilities are aimed at accelerating Chinese patient access to innovative medicines to address significant unmet need in AstraZeneca’s main therapy areas – respiratory; cardiovascular and metabolic diseases; and oncology. AstraZeneca’s commitment to bring cutting-edge biopharmaceutical science to China and to partner with the local science community is aligned with the Chinese Government’s focus on increasing innovation to support economic development and access to healthcare.

“AZD3759 is an example of China innovation for the global market. Asia iMed is on AstraZeneca’s innovation map.” Pascal Soriot, CEO

The initiatives and investments include: – An investment of $50 million to build an additional development and launch facility alongside our existing manufacturing site in Wuxi City to support the development and manufacture of innovative small molecules discovered in China and our global R&D sites – Additional investments include the creation of a new global hub for Pharmaceutical Development – alongside those in the UK and Sweden – with up to 50 scientists based in Shanghai and Wuxi City, to support both China and global needs. AstraZeneca is also establishing an integrated China medicines development organisation, bringing together early- and late-stage medicines development across small molecules and biologics – A strategic alliance with WuXi AppTec, a leading Chinese biologics manufacturer and contract research organisation, to produce innovative biologics locally in China – A strategic discovery partnership with Pharmaron, a leading R&D service provider based in China. Pharmaron works closely with our teams to deliver discovery services in chemistry, as well as in drug metabolism and pharmacokinetics (DMPK)

80 ©AstraZeneca 2016

The next wave of scientific innovation 81

An environment where science thrives

In December 2015, AstraZeneca, along with MedImmune, its global biologics research and development arm, announced a range of strategic initiatives to accelerate the delivery of medicines to patients in China, the company’s second largest market globally and a key growth platform.

clinical development activities, including all clinical development phases, and further developing local capabilities in drug substance synthesis and drug product development.

Collaborating for science innovation

Xiaolin Zhang, VP Asia iMed

AZD3759 is the first investigational drug discovered by Asia iMed and targets EGFR mutation positive advanced stage non-small cell lung cancer. – Clinical activities have been demonstrated in patients with CNS metastasis. Patients who have failed multiple lines of therapies showed benefit from AZD3759 treatment

IMED functions

“We have advanced our oncology portfolio significantly in 2015. Building on our expertise from AZD3759, we have identified the drug candidates that can penetrate the blood-brain barrier for the treatment of tumours that have metastasised to the brain.”

Above Xiaolin Zhang, VP Asia iMed

Therapy area progress

One of our strategic objectives is to realise the benefit of our new drugs for Chinese and Asian patients as quickly as possible. Asia iMed worked closely with global teams during 2015, and actively explored new potential drugs for the treatment of diseases that are most prevalent in China and Asia. Building on our pre-clinical findings, the olaparib team

has initiated AstraZeneca’s first Phase III study in gastric cancer. To ensure clinical studies in China are efficiently executed, the biomarker team also delivered essential biomarker studies for osimertinib, olaparib, volitinib, and MEDI4736 in 2015.

Introduction

Our drug discovery programmes in the chronic kidney disease area are progressing as planned. We prioritise kidney disease targets with a strong human genetic link, followed by animal models to establish causal relationship between the genetic change and disease phenotype. Realising the huge and increasing unmet medical needs, especially in China, we started our investment in the respiratory disease area, with a focus on chronic obstructive pulmonary disease (COPD).

Since entering China in 1993, AstraZeneca has been committed to continuously following the science, focusing on innovation and becoming one of the most trusted healthcare partners in improving the lives of Chinese patients. AstraZeneca’s China headquarters are based in Shanghai, and the company has more than 11,000 employees throughout the country. We’ve established manufacturing sites in Wuxi and Taizhou, as well as a China Distribution Centre in Wuxi. AstraZeneca also has an Innovation Centre in Shanghai, focused on the discovery and development of innovative candidate drugs to address the unique needs of patients in Asia.

Case study

Vanderbilt Center for Neuroscience Drug Discovery and AstraZeneca

The focus of our collaboration with the Vanderbilt Center for Neuroscience Drug Discovery (VCNDD) is to discover and develop novel positive allosteric modulators of the muscarinic M4 receptor for the treatment of psychiatric complications associated with Alzheimer’s disease and Parkinson’s disease.

Case study

Partnering to develop new medicines for neurodegenerative diseases

In 2013, AstraZeneca IMED entered into a collaboration with the Vanderbilt Center for Neuroscience Drug Discovery (VCNDD). Together, we hope to enable breakthrough discoveries and bring new medicines to patients who are suffering from neurodegenerative diseases.

Our collaboration is breaking new ground, with a new way of targeting this class of receptors where other efforts have been unsuccessful. Professor Jeffrey Conn, Director, Vanderbilt Center for Neuroscience Drug Discovery: “This new model for advancing neuroscience drug discovery pioneered by AstraZeneca fits perfectly with the mission of the VCNDD and makes it an ideal partnership for having an impact on these devastating disorders.

Opposite Neural network Above Professor Jeffrey Conn

This is a really special collaboration on multiple levels. First of all the science is innovative, and a fundamentally new approach to treatment of Alzheimer’s disease or other related neurodegenerative disorders. In diseases like Alzheimer’s and Parkinson’s, we still have a huge unmet medical need. There are very, very poor treatments available for patients and especially for the psychiatric complications, which can become very burdensome not only to the patients but also to the caregivers. In interacting with AstraZeneca as a scientist, it’s clear that they are there for a purpose, that they want to have an impact on patient care. They can see the possibilities of really having a positive impact. The thing I enjoy most about working with AstraZeneca is the shared passion. When we have meetings, when we talk on the phone, we can sense that passion and it creates an atmosphere where we’re very strategic, very focused. We get to the issues that are most important for the programme rather than thinking about process or other issues that could be distracting. I’m really excited to be a part of something as innovative as the Neuroscience iMed, discovering new treatments for neurosciencerelated disorders. Together we are aiming to get a new compound into clinical testing. Working together, we can explore new possibilities for treating patients who suffer from these devastating diseases.”

“Together with AstraZeneca we are aiming to get a new compound into clinical testing. Working together, we can explore new possibilities for treating patients who suffer from these devastating diseases.” Professor Jeffrey Conn, Director, Vanderbilt Center for Neuroscience Drug Discovery (VCNDD)

82 ©AstraZeneca 2016

The next wave of scientific innovation 83

Three science units with one shared goal Leveraging the combined power of AstraZeneca science

Discovery and Early Development

Our IMED Biotech Unit works together with colleagues in our Biologics team at MedImmune, and with Global Medicines Development, to discover and develop medicines that meet unmet medical needs.

Small Molecules Innovative Medicines and Early Development (IMED) Biotech Unit

By following the science, we are confident that together we can transform the lives of patients around the world.

Collaborations and Combinations

Late-stage Development

Introduction

AstraZeneca is a global, innovation-driven biopharmaceutical company that spans discovery, development, manufacturing, distribution and worldwide commercialisation. Our science exploits our rare combination of capabilities in small molecules and biologics, immunotherapies, protein engineering technologies and devices.

Combining the strength of our science units

Global Medicines Development

Market

MedImmune

Global Medicines Development

For further information please click here

For further information please click here

For further information please click here

Focuses on using state-of-theart discovery platforms and translational science in small molecules, oligonucleotides and other emerging technologies.

Focuses on biologics across our core areas and pioneers innovative research using unparalleled expertise in protein engineering, translational sciences and immunology.

The science engine room that drives late-stage development of our innovative pipeline, transforming exciting science into valued new medicines and ensuring patients around the world can access them.

Collaborating to exploit combination therapeutic strategies

Following the science, it became evident to the IMED team that both AZD9150 and AZD5069 inhibit signalling that tumours use to evade the host immune system. Paul Lyne, Senior Director and Global Project Lead for AZD9150 and AZD5069 believes that the combination of a Tumour Microenvironment (TME) modulator with immune checkpoint blockade offers a potential to improve patient outcomes as seen with immune checkpoint inhibition alone.

Carl Cook, Senior Director, Oncology TMU, IMED describes how working across the science units was, and will continue to be, critical to the accelerated progression of this project. “I believe we successfully achieved the first study milestone of this novel combination strategy as a result of a dynamic, collaborative team focused on the science and on patients. The IMED team really embedded themselves into the durvalumab team in GMD, and because of this we have not only been able to accelerate the programme, but to really drive synergy and efficiency. By working smartly, transparently and collaboratively, we manage a complex stakeholder network of senior leaders across the entire organisation. We’ve been able to create an environment where decision-making on the project is very effective and efficient. This approach, where we collaborate as one small team to drive communication and decision-making across other business units, translates into rapid decisionmaking, data sharing and best practices with the goal to accelerate the development of a potential novel combination strategy with durvalumab – to ultimately help more patients.”

“Immune checkpoint blockade therapies are transforming the therapeutic landscape for oncology patients, and have validated the clinical strategy of supporting the patient’s immune system to treat cancer. The next generation of immuno-oncology approaches will include therapies that combine complementary immune targeting mechanisms to

84 ©AstraZeneca 2016

The next wave of scientific innovation 85

An environment where science thrives

This important study milestone was successfully achieved dueto the collaborative approach, led by the AZD9150 team together with colleagues from the IMED, Global Medicines Development (GMD) and MedImmune organisations. Durvalumab, our Phase III PD-L1 checkpoint inhibitor, is currently demonstrating strong potential to combine with both immunotherapy and small molecules. We have an extensive development programme under way across our science units and across multiple tumour types and stages of disease, assessing the potential for immunotherapy to either replace or combine with traditional chemotherapy.

increase the proportion of patients that can benefit. These studies represent a key component of our organisation’s strategy for immuno-oncology and will hopefully bring increased benefit to patients”. Collaborating for science innovation

PD-L1 (MEDI4736)/AZD5069 and AZD9150 On 10 August 2015, the first patients were dosed on the durvalumab (MEDI4736) oncology combination trial with the oligonucleotide STAT3 inhibitor, AZD9150 or the small molecule CXCR2 inhibitor, AZD5069.

IMED functions

IMED Biotech Unit

Therapy area progress

Biologics MedImmune Biotech Unit

Collaborating and sharing data to redefine the future of drug discovery

In 2015, our teams established around 60 major collaborations covering key therapy areas and exciting new technologies that are set to drive progress in medical science innovation for years to come.

– Working side-by-side with external scientists to better understand disease mechanisms – Sharing information, expertise and insight – Making our compounds available through Open Innovation initiatives

“We have fruitful, illuminating exchanges of ideas and experience and expertise. We constantly exchange with AstraZeneca our results and try to subject them to discussion and we always receive valuable information from other AstraZeneca research groups. And this ecosystem makes things efficient and progressive.” Evgeny Imyanitov, Petrov Institute “The experience we are gaining is unmatched, and the contribution we can make is exciting. Our scientists are motivated by the innovation and the culture of sharing the best practice that AstraZeneca promotes.” Katherine Lee, Pharmaron “As a collaboration partner AstraZeneca is different from other pharma partners I have worked with in the past. They’re more willing to share information and we have found this across several programmes.” Jon Moore, Horizon

Several innovative compound-sharing agreements underlined our commitment to open innovation and information sharing. One of these involved a direct exchange of 210,000 compounds with Sanofi from our respective compound libraries. “We’ve worked hard to enrich our compound library in recent years and this exchange, which is by far the largest we’ve achieved, enables us to significantly increase its diversity. Most importantly, it will accelerate our ability to identify unique starting points that could become new medicines for patients,” said Mene Pangalos.

86 ©AstraZeneca 2016

The next wave of scientific innovation 87

An environment where science thrives

We joined a public-private consortium with Genomics England to accelerate the development of new diagnostics and treatments arising from the 100,000 Genomes Project. The GENE Consortium is a unique partnership between industry, academia and the National Health Service (NHS) Genomic Medicine Centres, which aims to transform treatment for patients with cancer and rare diseases, providing faster access to the right therapy and personalised healthcare.

– Sharing compounds to uncover novel target opportunities

Collaborating for science innovation

We engaged in a number of collaborations designed to advance treatment in areas of diabetes and chronic kidney disease, with key partners such as the University of Michigan, the French National Institute of Health and Medical Research (Inserm) and Harvard Stem Cell Institute. We also kicked off a partnership with the Montreal Heart Institute, which is now genotyping up to 80,000 DNA samples from our biobank, looking for genes associated with cardiovascular diseases and diabetes, their complications and treatment outcomes.

– In-licensing new chemical modalities and platforms

IMED functions

We signed four research collaborations aimed at harnessing the power of CRISPR, a pioneering genome-editing technique, across our entire discovery platform. Partnerships with the Wellcome Trust Sanger Institute, the Innovative Genomics Initiative, Thermo Fisher Scientific and Broad Institute/Whitehead Institute complement our in‑house CRISPR programme.

We work flexibly with our partners in pursuit of breakthrough science Therapy area progress

 ene Pangalos, Executive Vice President, M IMED Biotech Unit, on exchange of 210,000 compounds with Sanofi.

Partnering: a way of life

Introduction

“We’ve worked hard to enrich our compound library in recent years and this exchange, which is by far the largest we’ve achieved, enables us to significantly increase its diversity. Most importantly, it will accelerate our ability to identify unique starting points that could become new medicines for patients.”

Our teams are leading the way in creating open research environments that go beyond the usual collaboration models. We are always on the lookout for novel ways of working with others to advance medical science and speed up delivery of new medicines to patients. In 2015, we continued to build our network of collaborations with academic institutions, biotech and pharmaceutical companies in our key therapy areas as well as in rapidly evolving technologies such as CRISPR and antisense oligonucleotides. We are also pioneering new approaches to open innovation, creating a permeable research environment where scientists both inside and outside AstraZeneca can more freely share their ideas and collaborate on projects.

Global collaborations

A global science network UK 300

Sweden 100 Europe 160

Introduction

US 340

Our open innovation partnerships with academic translational drug discovery centres and governmentlinked funding agencies includes leading scientific institutions globally, who help facilitate our interactions with leading scientists. Our partners include: – United Kingdom: Medical Research Council

Asia 80

Advancing the science through Open Innovation Our Open Innovation initiative continued to gain momentum in 2015. Our Open Innovation portfolio now has around 24 clinical, 180 preclinical and 30 target innovation projects. We also added 30,000 new compounds to our high-throughput screening library and funded 12 R&D challenges during the year.

– Singapore: A*Star; National Health Innovation Centre-Duke

An invitation to innovate For further information please click here

Our Open Innovation portal makes it easy for external scientists to access our full range of Open Innovation programmes and find ways to advance medical science together. – Compound bank of ‘patientready’ active and discontinued compounds – Pharmacology toolbox of compounds with strong pharmacological properties – Collaborative effort to validate new targets, which may include high-throughput screening – Advanced cheminformatic capabilities to explore therapeutic potential of new molecules – R&D challenges open to anyone willing to offer innovative solutions

88 ©AstraZeneca 2016

Open Innovation initiative 24 clinical projects 180 preclinical projects 30 target innovation projects 30,000 new compounds added to our high-throughput screening library 12 R&D challenges funded during the year

An environment where science thrives

Out-licensing is another area of focus to ensure progression of indications that fall outside our core focus areas. In 2015, we signed deals with Millendo Therapeutics for AZD4901, an NK3 antagonist, for Polycystic Ovary Syndrome and Hot Flushes and with Corvidia for MEDI5117 (anti-IL-6 mAb), a precision medicine approach for Cardio-Renal Syndrome type 4.

– Taiwan: National Research Program for Biopharmaceuticals

Collaborating for science innovation

We also joined forces with the Wellcome Trust Sanger Institute, the European Bioinformatic Institute, Sage Bionetworks and the DREAM community on the AstraZeneca-Sanger Drug Combination Prediction DREAM Challenge, an established crowd-sourcing effort in the oncology area. Our unprecedented release of preclinical data from over 50 of our medicines reinforced our commitment to open innovation and our belief that therapeutic combinations have the potential to transform the way cancer is treated.

– Canada: NeoMed

IMED functions

With research facilities in a number of the world’s established and emerging scientific centres, we recognise the importance of leveraging our footprint to connect with the best external science, accelerating our scientific partnerships and alliances with leading academic and biotech partners around our sites as well as in other key locations across the globe.

– United States: National Institutes of Health/National Center for Advancing Translational Sciences; Academic Drug Discovery Network

Therapy area progress

Innovation without boundaries

– Germany: Lead Discovery Center

The next wave of scientific innovation 89

Case study

Ionis and AstraZeneca Partnering to develop the next generation of antisense-based therapeutics

Our most recent collaboration with US-based Ionis Pharmaceuticals, signed in August 2015, aims to discover and develop antisense therapies for cardiovascular, metabolic and renal diseases. This builds on a broad existing relationship and supports our strategic approach in these therapeutic areas using novel RNA-targeted treatments. Case study

Antisense drugs are short, chemically-modified, single-stranded nucleic acids (antisense oligonucleotides) that have the ability to target any gene product of interest. They offer new opportunities for therapeutic intervention because they act inside the cell to influence protein production by targeting RNA to either prevent the production of diseasecausing proteins, increase the production of proteins deficient in disease, or target toxic RNAs that are unable to generate proteins. Since our first collaboration with Ionis in 2012, we’ve continued to expand our partnership every year and are now working together in the key therapy areas of oncology, cardiovascular, metabolic and renal diseases. “We greatly value our collaboration with AstraZeneca. One aspect of the collaboration that we particularly value is the vision that AstraZeneca has for RNA therapeutics in general. They have placed a major investment in new platforms for drug discovery such as antisense, that go beyond the traditional drug platforms like small molecules and antibodies. We have been very pleased partnering with AstraZeneca over multiple collaborations. AstraZeneca has a strong vision for applying RNA therapeutic approaches to go after diseases that have significant unmet needs with current therapeutics on the market.” Brett Monia, SVP Drug Discovery, Ionis Pharmaceuticals “This expansion of our collaboration with AstraZeneca establishes our second strategic relationship. This new collaboration will help broaden the application of our antisense technology to targets in cardiovascular and metabolic disease. AstraZeneca is committed to finding novel bestin-class therapies for some of the largest, most complex and fastestgrowing disease segments in the developed world. Combining our antisense technology with AstraZeneca’s strong knowledge, leadership and commitment in these areas should be very valuable in fully exploiting these opportunities and moving new therapies effectively and efficiently toward the market.” B. Lynne Parshall, Chief Operating Officer at Ionis Pharmaceuticals “Antisense-based therapies are rapidly gaining momentum in the clinic and becoming an important component of our early-stage pipeline. Our collaborations with Ionis combine the world-class antisense drug research capabilities of Ionis with our expertise in oncology, cardiovascular and metabolic diseases drug discovery and development. By working together, we aim to uncover targets and pathways that can be manipulated using antisense drug therapy.” Mene Pangalos, Executive Vice President, IMED Biotech Unit

Above Ionis Pharmaceuticals, Carlsbad, California, US

90 ©AstraZeneca 2016

“AstraZeneca is committed to finding novel best-in-class therapies for some of the largest, most complex and fastest-growing disease segments in the developed world.” B. Lynne Parshall, Chief Operating Officer at Ionis Pharmaceuticals

The next wave of scientific innovation 91

A great place to work

Inspiring great scientists

We want to attract the brightest minds, the best young talent, the boldest innovators – people who share our passion for science and belief in the possible. In return, we offer a working environment that truly reflects our ambition to push the boundaries of science – a place where curiosity, innovation and collaboration flourish, where drive and determination is rewarded and where great science comes alive.

We also put continuous development of our people across IMED high on our agenda. From dedicated People Development Weeks to cross-team secondments and shadowing, our programmes ensure we continue developing the skills and capabilities to equip our scientists to be the best they can be. In quarter four of 2015 alone, we saw over ten IMED colleagues take up assignments outside their core role to broaden their learning and experience.

#31 Graduate Employer, The Guardian UK 300, 2014/2015.

Opposite CGI image of New Cambridge R&D Centre and Global HQ

Therapy area progress

In 2015, we welcomed more talented colleagues to our team, including accomplished scientists, respected academics and new graduates. They came for many reasons – the commitment to great science, the opportunity for personal development, the open culture, the inspiring values, the chance to be part of something life-changing. Whatever the reason, they have joined a truly great place to work.

In IMED, we’re committed to continually seeking ways to work across industry and academia to advance great science and address unmet patient needs. Some of our IMED colleagues come to us with long and distinguished academic careers – many of whom retain their academic links during their time with us – while others complement their AstraZeneca career by taking up teaching or research positions in the academic world. Our post-doc programme offers motivated, talented postdoctoral scientists the opportunity to make a difference with an academic-style position in a global pharmaceutical environment, and our graduate programme gives high-performing graduates the opportunity to gain experience across the research spectrum.

Introduction

Our commitment to scientific leadership rests on our ability to attract and retain the best scientists. Nowhere is this commitment more evident than in the way we recruit, develop and inspire our people.

! Bloomberg best employer 2016 #2

IMED functions

Stephen Rennard

Tim Eisen, PhD FRCP

VP, RIA iMed Adjunct Professor of Rheumatology at Gothenburg University

Chief Clinical Scientist, Clinical Discovery Unit, Early Clinical Development, AstraZeneca Richard and Margaret Larson Professor of Pulmonary Research, University of Nebraska Medical Center, Omaha, US

VP Head of Clinical Discovery Unit, Early Clinical Development & VP Interim Head of Oncology Translational Medicine Unit, Early Clinical Development, AstraZeneca Professor of Medical Oncology, University of Cambridge

92 ©AstraZeneca 2016

“My main interest as a respiratory physician is chronic obstructive pulmonary disease (COPD), which despite being a major cause of death worldwide is poorly understood and researched. The attraction for me in joining AstraZeneca was that the company is making a major commitment to respiratory disease and to driving the science. Novel treatments require novel approaches and AstraZeneca’s willingness to pursue these approaches offers the potential to impact drug development and clinical care. This role was an opportunity to participate in that. AstraZeneca has a lot of committed, hard-working scientists and there is every reason to believe that the drive to science in the company will lead to major advances.”

“At AstraZeneca I am working with a much broader range of people than I would do in academia. The IMED is an exciting place to work, where you can combine very good science with an ability to drive things forward. Things move much more quickly in AstraZeneca than in academia, so I can make faster progress on research that could impact patients in AstraZeneca than I could as a purely clinical academic. I am hoping to create a more productive relationship between pharma and academia and having experience of drug development gives me a better feel for the way industry works and where there are opportunities for collaboration. We in AstraZeneca spend an enormous amount of time developing talent. I think we offer young scientists a very active and accelerated career, with the training and opportunities to develop in industry and academia. It is an experience and an opportunity which I think is unique.”

The next wave of scientific innovation 93

An environment where science thrives

“Throughout AstraZeneca, there is true intent to follow the science. Combining my academic and clinical work with my AstraZeneca role puts me at the hard face of drug development. I see my primary job as being part of the IMED and driving our portfolio, but I have a live interface with patients and academia which keeps me awake and sharp, and which gives me many ideas. At AstraZeneca you can do world-class science but you can explore your scientific talents as well as your leadership talents. It’s a friendly, collaborative company and there is such a wide spread of people, skills and experiences. AstraZeneca offers an opportunity for curious people to explore.”

Collaborating for science innovation

Prof. Dr Maarten Kraan MD PhD

Björn Over

Katerina Pardali

Chief Scientist, CVMD iMed Professor of Nephrology and Physiology (St Peter’s Chair) at University College London

Postdoc, CVMD iMed, Medicinal Chemistry

Successfully completed the 2015 women in leadership programme

94 ©AstraZeneca 2016

An environment where science thrives

“I like the diversity at AstraZeneca and the flexibility I have to develop my skills. It’s such a pleasure to work with so many people who are willing to share expertise and ideas. What has impressed me most is the way the scientists work together. They try to find solutions and combine their expertise to get a better outcome. I’m very grateful for the experience to work in AstraZeneca, it’s been a great journey and I’ve learned a lot. I am not worried about my future now as I have gained so much experience on this programme.”

Collaborating for science innovation

IMED Graduate Scientist

In the AstraZeneca IMED we are committed to increasing the number of women in senior scientific roles. We firmly believe that the most innovative science is produced in diverse teams with different backgrounds, experiences and skills. That’s why we consistently seek to identify and develop the very best talent, wherever it exists. Our ‘Women as Leaders’ programme gives our female scientists a chance to come together to discuss issues such as career progression and personal development with a view to increasing their awareness of opportunities and the confidence to pursue them. We believe that by giving women the skills and support to make good career choices early, we will develop more role models and increase the number of women in senior roles... ultimately broadening diversity and driving innovation. In the 18 months the ‘Women as Leaders’ programme has been running, we have seen 30% of the participants take up expanded, larger roles.

IMED functions

Marta Wylot

Women as Leaders

Tim Eisen, VP Head of Clinical Discovery Unit, Early Clinical Development & VP Interim Head of Oncology Translational Medicine Unit, Early Clinical Development

Therapy area progress

“Having never had any experience of industry before, I’ve been very impressed and excited to meet the breadth of scientists in AstraZeneca. My academic and clinical colleagues are very interested, surprised and even envious of the opportunity I have had to be able to combine industry and academia and bring the two worlds together. I think the scientific ethos at AstraZeneca is very strong and the company has a reputation for being very sciencedriven. I think it’s an attractive option to consider for young scientists and for clinical scientists in particular. It is very hands-on, educational and stimulating. I still see patients in my clinic once a week and I get ideas from them that I can bring back into drug development programmes. I also continue to collaborate with colleagues in my clinical and academic roles but I can now bring added insight into the conversations.”

“The culture is really collaborative here. You have experts in so many fields, everybody is very supportive and they help each other out. People are curious about science; they love what they do and are really engaged. Without the guidance of the AstraZeneca experts, my project would not have been so successful. What we do as postdocs is really appreciated. We interact with academia and are able to publish our results, which is really important to our careers.”

Introduction

Robert Unwin

“The IMED is an exciting place to work, where you can combine very good science with an ability to drive things forward.”

The next wave of scientific innovation 95

Our strategic science centres In 2013, AstraZeneca announced plans to move our UK research activities to a new $500m facility in the centre of Cambridge. Our new facility at the Cambridge Biomedical Campus will become the company’s largest centre for oncology research and a centre of excellence for pre-clinical research, medicinal chemistry and high-throughput screening.

Our strategic R&D centre in Gothenburg is the centre of our research for two of our therapy areas; cardiovascular & metabolic diseases and respiratory & inflammation. It is also home to a large number of our scientists from our early phase Discovery Sciences unit and our Drug Safety and Metabolism team.

During 2015, we laid the foundations for our new home on the Cambridge Biomedical Campus, and expanded our interim high-quality lab and office facilities to accommodate our growing presence in the city.

Our vibrant Gothenburg facility has seen the BioVentureHub go from strength to strengthh since its inception in 2014, with 14 companies and one academic group now working in this innovative ecosystem.

Our evolving science footprint in the North West means a small number of IMED colleagues remain at our Macclesfield campus, and at Alderley Park until our R&D exit of the site is completed. The growing Alderley Park BioHub is successfully creating an optimum environment for emerging businesses to thrive.

Below AstraZeneca’s strategic R&D centre in Gothenburg

Launched in 2015, the Gothenburg ‘Coffee Lab’ is an AstraZeneca first – to inspire employees to world-class ideas development.

Opposite top left AstraZeneca’s smallmolecule research facility in Boston, North America

External designers worked with a project team of employees from different functions across the site to create a creative space for meetings, socialising and relaxation.

Stimulating ‘cross-fertilisation’, both between the hub companies and with AstraZeneca, is key to the success of the biohub approach in our evolving sites.

Opposite bottom left AstraZeneca’s smallmolecule research facility in Shanghai, China Opposite bottom right The Gothenburg ‘Coffee Lab’ is an AstraZeneca first – to inspire employees to world-class idea development

US – Boston

The best innovative ideas often come out of informal chats rather than formal meetings, so the site is creating an informal meeting point, where colleagues can spend time away from a desk or meeting room boundaries.” Jenny Sundqvist, Site Director

“AstraZeneca has been on a transformative journey over the past few years, placing great science at the heart of everything we do in the delivery of breakthrough medicines to patients. Our ambition is to improve the lives of 200 million people by 2025. Such an ambition would not be possible without establishing collaborations of all types with academia and industry. Our biohubs provide a fantastic opportunity to explore collaboration even further.” Kumar Srinivasan, Head of AstraZeneca R&D Boston and VP Scientific Partnering and Alliances

IMED functions

Boston is home to AstraZeneca’s smallmolecule research in North America, with state-of-the-art laboratories in Waltham, just west of the city centre, and our Neuroscience team in the heart of the city’s Technology Square. Our Boston-based scientists focus on the discovery and development of new medicines for the treatment of cancers and neurological disorders. The site also houses the Gatehouse Park BioHub, which is thriving with nine research companies already in place since launch in September.

“The idea is to stimulate new ways of interacting with colleagues from functions that you usually don’t meet. Coffee Lab has built on the success of the existing lounge, which draws people from all over the site to get together for meetings and exchanges of ideas. The lounge is the place to meet, and we want to build on that buzz with a larger area for both scheduled and spontaneous meetings.

A bold, new R&D initiative to foster life sciences discovery and the exchange of ideas between scientists, our Gatehouse Park BioHub, along with the BioVentureHub at the Gothenburg site in Sweden, and the BioHub at our Alderley Park site in the UK, all have vibrant but distinctive features offering an energising environment, all about sharing ideas and tapping into great science.

Therapy area progress

Sweden – Gothenburg

Creating vibrant Biohubs

Introduction

UK – Cambridge

Our creative area to stimulate innovative thinking

Collaborating for science innovation An environment where science thrives

China – Shanghai Our small-molecule research facility in China is located at the Zhangjiang High Tech Park in the Pudong area of Shanghai. Our research teams here focus on discovering potential new medicines that meet the unique needs of patients in Asia and drive forward translational science across our core therapy areas.

96 ©AstraZeneca 2016

The next wave of scientific innovation 97

Building our future Our new Cambridge site

Our new R&D Centre will become the company’s largest centre for oncology research and a centre of excellence for pre-clinical research, medicinal chemistry and highthroughput screening. Beyond cancer research, our R&D will focus on cardiovascular and metabolic diseases, respiratory, inflammation and autoimmune diseases and conditions of the central nervous system.

In 2015, we laid the foundations for our new home on the Cambridge Biomedical Campus. This proximity to leading research and academic institutions is key to our culture of open innovation and partnering. We also continue to develop scientific partnerships and outreach programmes. While these are supported by our local presence, they have UK-wide and global reach.

Science in pictures

98 ©AstraZeneca 2016

Supporting the next generation of scientists In 2015, we began funding academic clinical lectureships and PhD students at the University of Cambridge, with 80 agreed across AstraZeneca and MedImmune over the next five years. We also support the Cambridge Judge Business School’s “Accelerate” programme designed to identify, train and mentor start-up life science businesses. AstraZeneca’s volunteer mentors come from a range of roles, with expertise in areas like business development, intellectual property and innovation alliances. Cambridge Cancer Science Symposium As part of our open innovation strategy, IMED and MedImmune brought top scientists from academia and industry together to share the next generation of oncology science at the Cambridge Cancer Science Symposium, Churchill College. Delegates were not only impressed by the quality of the science, but also the opportunity to get so many academic and industry organisations together under one roof, sharing different perspectives in striving for the same goal – to accelerate new and improved treatment options for cancer patients.

An environment where science thrives

In addition, our PhD programmes with the University of Cambridge and ongoing commitment to STEM programmes in the local community underline our commitment to support, develop and inspire the next generation of Cambridge scientists. By moving to Cambridge, AstraZeneca is helping to build an attractive UK life sciences destination for investment, and deliver a magnet for top scientific talent – underpinned by a world-leading science base, a vibrant and entrepreneurial environment that drives innovation, as well as timely patient access to innovative medicines. Cambridge Biomedical Campus will be an open, welcoming and vibrant centre that will inspire our IMED team and our partners to push the boundaries of scientific innovation.

Science retreat In October, we welcomed over 250 IMED scientists to our Science Retreat at Robinson College in Cambridge. Opened by Nobel Laureate Sir Venki Ramakrishnan, the meeting immersed delegates in inspiring science with topics ranging from immunology, to our progress in open innovation, the patient perspective plus a futuristic look into science and technology that may impact IMED research over the next decade.

Collaborating for science innovation

Our new Cambridge site will house 2,300 colleagues, with world-class capabilities in target biology, medicinal chemistry, protein engineering, translational science, biopharmaceutical and clinical development. Our presence in Cambridge allows us to play an active role in enabling a permeable scientific hub, where the best ideas flow out, as well as in.

IMED continued integration into the Cambridge community during 2015

IMED functions

At the end of 2015, we welcomed our thousandth AstraZeneca employee into Cambridge. This strong and growing presence allows us to deepen our scientific relationships as part of the local life-sciences ecosystem, before we move into our new R&D Centre where IMED MedImmune and Global Medicines Development will sit side by side in an open, collaborative workplace. The site will bring together our small molecule and biologics R&D, as well as all our discovery science capabilities and late-stage development, opening up opportunities to work collaboratively across these areas to create the next generation of medicines that will positively impact the lives of millions of people.

“Science in pictures”, is an artwork installation on the hoardings around the construction site of our new R&D Centre and Corporate Headquarters on the Cambridge Biomedical Campus. Following workshops led by AstraZeneca and MedImmune scientists and local artists in the summer, 400 students at nine local schools were invited to create a circular picture that captures what science means to them. These workshops provided an insight into the science behind new medicines and how potential medicines are identified, and developed. In addition to active volunteer-led programmes in schools, AstraZeneca and MedImmune also sponsor outreach initiatives through the Cambridge Science Festival, Big Biology Day and the Cambridge Science Centre. This installation is an expression of our commitment to community outreach through science education and will be visible to the Cambridge Biomedical Campus community and its visitors throughout 2016.

Therapy area progress

It’s an exciting time for IMED as we continue to establish ourselves in the Cambridge science community, building on the long-standing presence of our colleagues from AstraZeneca’s global biologics research and development arm, MedImmune. We chose to be in Cambridge because we wanted to be at the heart of one of the best scientific centres in the world.

Introduction

In 2013, AstraZeneca announced plans to build a global Research and Development Centre and its Corporate Headquarters on the Cambridge Biomedical Campus. This is one of our flagship initiatives and is part of redefining our future and aspiration to be one of the best scientific institutions in the UK and globally.

Opposite and top left CGI images of AstraZeneca’s new Global R&D Centre and Corporate Headquarters, Cambridge, UK

The next wave of scientific innovation 99

Case study

The next wave of innovation in DNA Damage Response

Targeted therapy based on inhibiting the DNA Damage Response (DDR) in cancers offers the potential for a greater therapeutic window by tailoring treatment to patients

There are three main aspects of the DDR that are different in cancer and therefore provide a rationale for drug targeting: – DDR pathway loss results in greater dependency on remaining DDR pathways – Increased replication stress leads to greater dependency on ATR-CHK1-Wee1 – Increased levels of endogenous damage and genomic instability results in greater sensitivity to exogenous DNA damage

Targeting DDR in cancer An underlying hallmark of cancers is their genomic instability, which is associated with a greater propensity to accumulate DNA damage. Historical treatment of cancer by radiotherapy and DNA-damaging chemotherapy is based on this principle, yet it is accompanied by significant collateral damage to normal tissue and unwanted side effects. Targeted therapy based on inhibiting the DNA Damage Response (DDR) in cancers offers the potential for a greater therapeutic window by tailoring treatment to patients with tumours lacking specific DDR functions. An invited review in Molecular Cell by Mark O’Connor (Oncology iMed) summarises the scientific data behind olaparib in the context of it being the first approved, targeted cancer medicine for patients with a tumourspecific deficiency in their DDR biology, and it discusses the future significance of DDR-based agents in cancer therapy. Covering all DDR-targeted agents that either have been approved or are in clinical development, the article demonstrates that AstraZeneca has a world-leading pipeline of compounds that target DDR pathways in cancer.

AstraZeneca portfolio targets distinct aspects of DDR Chosen for both their different roles in DNA repair and when in the cell cycle they play their primary role

Phase I AZD0156 Effect is manifest in M phase

AZD2811 Effect is manifest in M phase

M S

Target: ATM Effect: Inhibits repair of Double Strand Breaks

100 ©AstraZeneca 2016

Effect is manifest in M phase

M G1

G2

AZD6738

Phase III, approved

AZD1775

Olaparib

Effect is manifest in M phase

M G1

G2

Phase II

S

Target: AURORA B Effect: Deregulation of chromosome segregation and cytokinesis

M G1

G2

Effect is manifest in M phase

S

Target: ATR Effect: inhibits S phase replication stress response and repair of Double Strand Breaks

M G1

G2 S

G1

G2 S

Target: Wee1 Effect: inhibits S phase replication stress response and G2/M cell cycle checkpoint

Target: PARP Effect: inhibits repair of Single Strand Breaks

The next wave of scientific innovation 101

Case study

Building a world-leading pipeline

The AstraZeneca portfolio targets distinct aspects of the DNA Damage Response (DDR). This relates both to their different roles in DNA repair and at what point in the cell cycle they exert their effect. AstraZeneca has a number of DDR-targeted compounds in clinical development.

The latest updates on development of AstraZeneca’s DDRtargeted compounds are listed: all ongoing olaparib Phase III trials (targeting PARP), AZD1775 (Wee1) Phase II trials and AZD6738 (ATR) Phase I trials are included. AZD0156 (ATM) has also entered clinical trials.

Olaparib – the first medicine based on DDR The recent approval of olaparib the poly (ADP-ribose) polymerase (PARP) inhibitor for treating tumours harbouring BRCA1 or BRCA2 mutations, represents the first medicine based on this principle, exploiting an underlying cause of tumour formation that also represents an Achilles’ heel.

Strategy for the use of DDR inhibitors as anti-cancer agents

Effect is manifest in M phase

G1

G2/M checkpoint Allows time to repair any remaining DNA DSBs before attempting cell division DDR targets: CHK1, MYT1, Wee1

There are potential advantages of combining olaparib with other DDR-targeted compounds, or with agents such as the AstraZeneca VEGFR TKI cediranib that target other cancer pathways; these combinations could provide broader and more effective responses than a monotherapy approach.

M PREVENT REPAIR

G2

DNA replication phase

G2

Gap/growth Phase II

M

Cell division phase

\

Cell cycle checkpoint

G1 S MAXIMIZE DAMAGE

S-phase checkpoint Delays replication process to allow time to deal with unrepaired DNA damage or DNA damage resulting from collapsed replication forks DDR targets: ATR, CHK1, DNA-PK, Wee1

DNA replication stress – a promising target for DDR-based therapies Another hallmark of cancer linked to the DDR is DNA replication stress, which occurs to a greater degree in cancer cells than normal cells and is therefore a potential target for DDR-based therapies such as AZD6738 and AZD1775, which inhibit the DDR regulators ATR and Wee1, respectively. DNA damage caused during the DNA replication phase of the cell cycle (S phase) can lead to cell death if it is not repaired before cell division (M phase): one therapeutic strategy to maximize the amount of DNA damage is to inhibit the checkpoints at which the cell cycle is halted until any DNA damage has been repaired; for example, AZD1775 inhibition of Wee1, which regulates the G2/M checkpoint, allows accumulated DNA damage to be carried into M phase, inducing cancer cell death.

Mark O'Connor, Senior Principal Scientist IMED Biotech Unit (Oncology)

Gap/growth Phase I

S

Investigation of both the cell cycle and cell death effects resulting from treatment with the ATR and Wee1 inhibitors in DLBCL models highlighted differences consistent with the greater potency of the Wee1 inhibitor in these models; an assessment of in vivo activity further supported these findings. Results presented at the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutic in November on AZD1775 treatment of a larger panel of in vivo patient-derived explant (PDX) models of multiple tumour types demonstrated both the significant breadth and depth of the Wee1 inhibitor single-agent activity. There is also potential for this activity to be enhanced further through combination with olaparib, a PARP inhibitor that induces S-phase DNA damage. Together, these data have led to the recent initiation of AZD1775 monotherapy as well as AZD1775/ olaparib combination clinical trials.

G1/S checkpoint Allows time to repair DNA damage before starting DNA replication DDR targets: ATM, CHK2, p53

What’s next? The Phase III olaparib development programme also includes two additional studies: a prostate cancer study that received Late Storage Development Committee approval on 9 November 2015 (D081DC00007) and that has now achieved FDA breakthrough status. In addition, there is also a study of olaparib in combination with durvalumab (DUO study, D081KC00002). The ATM inhibitor AZD0156 is the latest DDR targeted agent to enter the clinic and will be used in combination with olaparib to investigate whether this novel DDR agent combination can extend the patient population that can benefit from olaparib. AZD0156 also has the potential to increase the effectiveness of chemotherapies that require ATM function. Pivotal to optimising the clinical use of this new therapeutic category of anti-cancer agents will be the selection of the most appropriate treatment combinations for DDR-targeted therapies. Due to the strong mechanistic links between DDR and different aspects of the immune response, this includes the potential for combinations with immunotherapy agents. Another key aspect for success will be the targeting of specific patient populations whose cancers carry definable DDR gene mutations. What is clear is that targeting DDR represents an exciting new advance in our ability to treat cancer.

1

O’Connor MJ. Targeting the DNA damage response in cancer. Molecular Cell 2015; 60(4)

102 ©AstraZeneca 2016

The next wave of scientific innovation 103

Case study

By listing all the ongoing Phase III trials involving PARP inhibitors, it becomes clear that the AstraZeneca olaparib Phase III programme is far more extensive than that of any of the four other molecules in this class from other manufacturers that are under investigation in the clinic (niraparib, rucaparib, talazoparib and veliparib).

Selective targeting of tumours that harbour a DDR deficiency means that, unlike with radiotherapy or chemotherapy, tumour cells can be killed without causing significant side effects or damage to normal tissue. In 2015, clinical validation was provided by regulatory approval of the PARP inhibitor olaparib in ovarian cancer patients whose tumours have a mutation in BRCA1 or BRCA2, which encode proteins involved in the DDR. Olaparib activity is now being explored in the clinic in non-BRCA DDR deficient cancers (for example ATM-low gastric cancers in the Phase III Gold trial in Asian patients), and additionally in prostate cancer.

Our reputation for scientific leadership

Our continued drive to develop a thriving science environment has generated great progress during 2015, both inside IMED and within the broader scientific ecosystem.

Our publications We strengthened our scientific reputation through an increased focus on high-quality scientific publications in 2015 with 455 papers published. We also saw outstanding progress in publishing our science in high-impact, peerreviewed journals, moving from a single high-impact publication in 2010 to 29 in 2015.

c-kit+ cells do not generate lung epithelium during maintenance and repair

American Journal of Respiratory and Critical Care Medicine

Nature Communications

Nature Medicine

About the paper: The FGFR family of kinases are key mediators of both developmental and disease associated blood vessel growth. Prior work had only ever shown FGFR1 with a key element to binding ATP (the energy source) being folded in ready to activate the protein. This paper, a collaboration between IMED and MedImmune scientists, for the first time details the interactions and stability associated with protein being ‘flipped’ into a non-active form.

About the paper: It has been reported that c-kit+ progenitor cells resident in the human lung regenerate epithelial cells upon transplantation into injured mouse lung. For the first time, our scientists demonstrated that during normal function and regeneration conditions after injury, c-kit+ cells adopt vascular endothelial cell fate and not any type of lung epithelial cells. In addition, c-kit+ cells proliferate after injury and contribute to new blood vessel formation within the lung.

About the paper: Lymphoid follicles have been associated with COPD disease severity, with localised overexpression of B cell-activating factor (BAFF) demonstrated in patients with severe COPD. This paper, a collaboration between IMED, MedImmune and Ghent University Hospital has further described the role of BAFF in COPD, demonstrating BAFF overexpression in COPD patient lung tissue and in a mouse model of chronic cigarette smoke exposure. Furthermore, it was shown that antagonising BAFF can protect against alveolar destruction and pulmonary inflammation.

Lead AstraZeneca authors: Gareth Davies, Geoff Holdgate and Chris Phillips

Lead AstraZeneca authors: Anja Schinwald, Danen Cunoosamy, Claudie Malanda, Alan Sabirsh, Eileen McCall, Liz Flavell, Ronald Herbst

Impact: This study unravelled the true fate of c-kit+ cells during lung homeostasis and lung repair, calling attention to the clinical application of c-kit+ progenitor cells as lung epithelial progenitors for the treatment of pulmonary disease. Lead AstraZeneca author: Qing-Dong Wang

IMED Science Awards

The 2015 IMED Science Retreat took place in October at Robinson College, Cambridge. The packed agenda of scientific innovation, patient insight and technology of today and tomorrow helped deliver a meeting of high-quality science and inspiration, opened by Nobel-winning structural biologist Sir Venki Ramakrishnan. The IMED Science Retreat is an important event in the AstraZeneca science calendar, enabling colleagues to get a view of the latest developments outside their areas, share ideas and look at how we can apply science in new and different ways to drive scientific leadership.

Our continued progress towards scientific leadership is down to the collective effort of dedicated and talented individuals striving to make a difference. The prestigious annual IMED Science Awards aim to recognise and reward individual and team efforts, share great achievements and inspire even more great work from our scientists.

“What a fantastic and inspirational event, the way our scientists are pushing the boundaries of everything we do make me very proud.” Mene Pangalos, Executive Vice President, IMED Biotech Unit

104 ©AstraZeneca 2016

The next wave of scientific innovation 105

An environment where science thrives

“This environment, buzzing with science and energy, is a great place for learning about the research going on across IMED, and to share our progress. I really enjoyed the patient insight sessions, they were truly powerful and reminds us of our goal.” Sepideh Hagvall Heydarkhan, Associate Principal Scientist CVMD iMed

The 2015 IMED Science Awards celebrated some of the best breakthrough, high-impact science taking place at AstraZeneca. 150 global nominees were invited to join the IMED Leadership Team and members of the review panel at a black-tie celebratory dinner. The awards recognised outstanding scientific achievement; high impact work acknowledged as gamechanging. Winning teams and individuals received trophies and certificates, and are also rewarded with tailored opportunities to support future research and enhance their careers.

Collaborating for science innovation

Science Retreat

IMED functions

Impact: This research has demonstrated novel findings that will influence future strategies in the treatment of COPD.

Impact: This work has significance in the design of kinase inhibitors and the understanding of the stabilisation of the non-active form of the target protein.

Therapy area progress

Structural and dynamic insights into the energetics of activation loop rearrangement in FGFR1 kinase

Introduction

Our commitment to being a science-led company doesn’t end with our work in the lab. We believe that continued innovation relies on us fostering a culture of scientific excellence, empowering our scientists to not only keep abreast of the latest developments and breakthroughs, but to drive them.

Role of B Cell–Activating Factor in Chronic Obstructive Pulmonary Disease

High impact publications in 2015 Title

Authors

Publication

Title

Authors

Cancer Cell

Acetyl-coA synthetase 2 promotes acetate utilization and maintains cancer cell

Schug ZT, Peck B, Jones DT, Zhang Q, Grosskurth S, Alam IS, Goodwin LM, Smethurst E, Mason S, Blyth K, McGarry L, James D, Shanks E, Kalna G, Saunders RE, Jiang M, Howell M, Lassailly F, Thin MZ, Spencer-Dene B, Stamp G, van den Broek NJ, Mackay G, Bulusu V, Kamphorst JJ, Tardito S, Strachan D, Harris AL, Aboagye EO, Critchlow SE, Wakelam MJ, Schulze A, Gottlieb E

Nature Communications

Structural and dynamic insights into the energetics of activation loop rearrangement in FGFR1 kinase

Klein T, Vajpai N, Phillips J, Davies G, Holdgate G, Phillips C, Tucker J, Norman R, Scott A, Higazi D, Lowe D, Breeze A

Nature Medicine

Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M

Thress KS, Paweletz CP, Felip E, Cho BC, Stetson D, Dougherty B, Lai Z, Markovets A, Vivancos A, Kuang Y, Ercan D, Matthews SE, Cantarini M, Barrett JC, Jänne PA, Oxnard GR

Nature Medicine

c-kit+ cells do not generate lung epithelium during maintenance and repair

Liu Q, Huang X, Zhang H, Tian X, He L, Yang R, Yan Y, Wang Q, Gillich A, Zhou B

Cancer Cell

Nature Reviews Cancer

MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road

Caunt CJ, Sale MJ, Smith PD, Cook SJ

Cell Research

Genetic lineage tracing identifies in situ kit-expressing cardiomyocytes

Zhang WJ, Cristinacce P, Bondesson E, Nordenmark L, Young SS, Liu YZ, Singh D, Naish JH, Parker GJM

Nature Reviews Drug Discovery

An analysis of the attrition of drug candidates from four major pharmaceutical companies

Waring MJ, Arrowsmith J, Leach AR, Leeson PD, Mandrell S, Owen RM, Pairaudeau G, Pennie WD, Pickett SD, Wang J, Wallace O, Weir A

European Heart Journal

Effect of genetic variations on ticagrelor plasma levels and clinical outcomes

Varenhorst C, Eriksson N, Johansson A, Barratt BJ, Hagström E, Åkerblom A, Syvänen AC, Becker RC, James SK, Katus HA, Husted S, Steg G, Siegbahn A, Voora D, Teng R, Storey RF, Wallentin L

Nature Reviews Drug Discovery

ESKAPEing the labyrinth of antibacterial discovery

Tommasi R, Brown D, Walkup G, Manchester J, Miller A

Randomized, double-blind Phase II trial with prospective classification by ATM protein level to evaluate the efficacy and tolerability of olaparib plus paclitaxel in patients with recurrent or metastatic gastric cancer

Bang YJ, Im SA, Lee KW, Cho JY, Song EK, Lee KH, Kim YH, Park JO, Chun HG, Zang DY, Fielding A, Rowbottom J, Hodgson D, O’Connor MJ, Yin X, Kim WH

Nature Reviews Drug Discovery

Pioneering government-sponsored drug repositioning collaborations: progress and learning

Frail DE, Brady M, Escott J, Holt A, Sanganee HJ, Pangalos MN, Watkins C, Wegner CD

Nature Reviews Drug Discovery

Towards a hit for every target

Rees S, Gribbon P, Birmingham K, Janzen W, Pairaudeau G

Journal of Clinical Oncology

Molecular profiling and targeted therapy for advanced thoracic malignancies: a biomarkerderived, multiarm, multihistology phase II basket trial

Lopez-Chavez A, Thomas A, Rajan A, Raffeld M, Morrow B, Kelly R, Carter CA, Guha U, Killian K, Lau CC, Abdullaev Z, Xi L, Pack S, Meltzer PS, Corless CL, Sandler A1, Beadling C, Warrick A, Liewehr DJ, Steinberg SM, Berman A, Doyle A, Szabo E, Wang Y, Giaccone G

Nature Reviews Drug Discovery

Therapy area heat map for emerging markets

Gautam A, Li L, Srinivasan K

Neuron Molecular Cell

Targeting the DNA damage response in cancer

O’Connor MJ

BrainSeq: neurogenomics to drive novel target discovery for neuropsychiatric disorders

Schubert C, O’Donnell P, Quan J, Wendland J, Hualin S, Domenici E, Essioux L, Kam-Thong T, Didriksen M, Matsumoto M, Saito T, Brandon N, Cross A, Wang Q, Heon Shin J, Jaffe A, Jia Y, Straub R,Deep-Soboslay A, Hyde T, Kleinman J, Weinberger D

Nature

Patient-centric trials for therapeutic development in precision oncology

Biankin AV, Piantadosi S, Hollingsworth SJ

New England Journal of Medicine

AZD9291 in EGFR inhibitor–resistant non–small-cell lung cancer

Jänne PA, Yang JC, Kim DW, Planchard D, Ohe Y, Ramalingam SS, Ahn MJ, Kim SW, Su WC, Horn L, Haggstrom D, Felip E, Kim JH, Frewer P, Cantarini M, Brown KH, Dickinson PA, Ghiorghiu S, Ranson M

Nature

Precision medicine: AstraZeneca’s approach

March R

Science

A sustainable model for antibiotics

Perros M

Nature Chemical Biology

Translating slow-binding inhibition kinetics into cellular and in vivo effects

Walkup G, You Z, Ross P, Allen E, Daryaee F, Hale M, O’Donnell J, Ehmann D, Schuck V, Buurman E, Choy A, Hajec L, MurphyBenenata K, Marone V, A Patey S, Grosser L, Johnstone M, Walker S,Tonge P, Fisher S

Science Advances

Structural basis of lewisb antigen binding by the helicobacter pylori adhesin BabA

Nage N, Howard T, Phillips C, Brassington C, Overman R, Debreczeni J, Gellert P, Stolnik G, Winkler G, Falcone F

Triaminopyrimidine is a fast-killing and long-acting antimalarial clinical candidate

Hameed S, Solapure S, Patil V, Henrich P, Magistrado P, Bharath S, Murugan K, Viswanath P, Puttur J, Srivastava A, Bellale E, Panduga V, Shanbag G, Awasthy D, Landge S + et al..

Science Translational Medicine

AZD9150, a Next-Generation Antisense Oligonucleotide Inhibitor of STAT3, with Early Evidence of Clinical Activity in Lymphoma and Lung Cancer

Aberrant splicing of U12-type introns is the hallmark of ZRSR2 mutant myelodysplastic syndrome

Madan V, Kanojia D, Li J, Okamoto R, Sato-Otsubo A, Kohlmann A, Sanada M, Grossmann V, Sundaresan J, Shiraishi Y, Miyano S, Thol F, Ganser A, Yang H, Haferlach T, Ogawa S, Koeffler P

Hong D, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, Fowler N, Zhou T, Schmidt J, Jo M, Lee SJ, Yamashita M, Hughes SG, Fayad L, Piha-Paul S, Nadella MVP, Mohseni M, Lawson D, Reimer C, Blakey DC, Xiao X, Hsu J, Revenko A, Monia BP, MacLeod AR

Science Translational Medicine

Tissue transcriptome-driven identification of epidermal growth factor as a chronic kidney disease biomarker

Ju W, Nair V, Smith S, Zhu L, Shedden K, Song PX, Mariani LH, Eichinger FH, Berthier CC, Randolph A, Lai JY, Zhou Y, Hawkins JJ, Bitzer M, Sampson MG, Thier M, Solier C, Duran-Pacheco GC, Duchateau-Nguyen G, Essioux L, Schott B, Formentini I, Magnone MC, Bobadilla M, Cohen CD, Bagnasco SM, Barisoni L, Lv J, Zhang H, Wang HY, Brosius FC, Gadegbeku CA, Kretzler M; ERCB, C-PROBE, NEPTUNE, and PKU-IgAN Consortium

Journal of Clinical Oncology

Nature Communications

Nature Communications

Nature Communications

106 ©AstraZeneca 2016

Oxidation of the alarmin IL-33 regulates ST2-dependent inflammation

Cohen S, Scott I, Majithiya J, Rapley L, Kemp B, England E, Rees G, Overed-Sayer C, Woods J, Bond N, Seguy-Veyssier C, Embrey K, Sims D, Snaith M, Vousden K, Strain M, Chan D, Carmen S, Huntington C, Flavell L, Xu J, Popovic B, Brightling C, Vaughan T, Butler R, Lowe D, Higazi D, Corkill D, May R, Sleeman M, Mustelin T

The next wave of scientific innovation 107

An environment where science thrives

Pommier AJ, Farren M, Patel B, Wappett M, Michopoulos F, Smith NR, Kendrew J, Frith J, Huby R, Eberlein C, Campbell H, Womack C, Smith PD, Robertson J, Morgan S, Critchlow SE, Barry ST

Collaborating for science innovation

Leptin, BMI and a metabolic gene expression signature associated with clinical outcome to VEGF inhibition in colorectal cancer

Cell Metabolism

IMED functions

Schwartz S, Wongvipat J, Trigwell CB, Hancox U, Carver BS, Rodrik-Outmezguine V, Will M, Yellen P, de Stanchina E, Baselga J, Scher HI, Barry ST, Sawyers CL, Chandarlapaty S, Rosen N

Therapy area progress

Feedback suppression of PI3Ka signalling in PTENmutated tumours is relieved by selective inhibition of PI3Kß

Introduction

Publication

Preparing for the future with our ‘IMED Futures’ teams

During 2015, our teams interrogated emerging technologies, explored new approaches to drug discovery and challenged conventional thinking in the search for new opportunities to bring benefit to patients. Here we shine a spotlight on four of our programmes.

Digital health

Targeted drug delivery

Current technology allows measuring the body’s vital signs using health patches but our team recognises the true value for pre-clinical monitoring lies with invasive biosensor technology. In the future, developing pre-clinical sensors that monitor drug exposure and biomarkers of safety and efficacy will have the potential for clinical use – providing online biosensors for patients, and hence changing the status quo for patient care. “Today we have already seen the approval of wearable devices to measure online glucose levels for diabetic patients. If we can discover reliable biomarkers and couple these with sensitive sensor technology this opens up a wealth of opportunities.” Celina d’Cruz, Pre-clinical Futures Lead

Microphysiological systems As we advance our understanding of biology and integrate our knowledge of cellular behaviour and tissue function, it is apparent that the current pre-clinical in vitro models have limitations when predicting organ functionality. Complex cellular microphysiological systems (MPS), consisting of interacting organs-on-chips or tissue-engineered, 3D organ constructs, present an opportunity to bring new tools to biology, medicine, pharmacology, physiology, and toxicology. By placing human or animal cells in a more ‘natural’ environment, we can start to recapitulate the dynamics of drug-organ, drug-drug, and drug-organ-organ interactions to allow better predictivity of clinical translation. Our team is collaborating with some of the leading experts in the world at TissUse, Harvard and Vanderbilt Universities.

“Connecting data stacks of patient data is not new. What would be cutting-edge is if we could design them to handle ever increasing data types.” Hitesh Sanganee, Digital Futures Lead

108 ©AstraZeneca 2016

The next wave of scientific innovation 109

An environment where science thrives

“With advances in cell culture and microfluidics it is now possible to emulate human biology on a microscale. Taking this one step further and connecting these organ units together, human-on-achip technology will allow us to advance our understanding of the safety and efficacy earlier in drug discovery.” Lorna Ewart, Microphysiological Systems Futures Lead

Collaborating for science innovation

Improved understanding of the physiological barriers to efficient drug delivery has resulted in significant advances in delivery systems. This, coupled with novel analytical and imaging techniques allow for even more sophisticated delivery systems opening up new target space. Our team is looking to improve target efficacy by enhancing our targeting capabilities to allow delivery of both small molecules and oligonucleotide therapeutics – miRNA, mRNA and antisense. To do this our team has focused on three key areas: 1) cellular targeting, 2) improving cellular uptake and 3) enhancing drug delivery. In the latter case, we have already initiated collaborations with BIND Therapeutics for their ACCURINS® polymeric nanoparticle technology and with Starpharma exploring their dendrimer technology platform. Both these technologies improve therapeutic index and ability to formulate challenging molecules.

To enhance the impact of current online data monitoring, our teams have been exploring the possibilities within the rich data source provided by sensor technology and wearable devices. Incorporating biosensor technology into our pre-clinical studies will allow us to change current practice, improve translation and safety read-outs, while reducing the number of animal studies.

IMED functions

“Connecting data stacks of patient data is not new. What would be cutting-edge is if we could design them to handle ever increasing data types, ‘stacking’ multiple layers of patient data – making them flexible to connect with whichever data is current.” Hitesh Sanganee, Digital Futures Lead

Pre-clinical futures

Therapy area progress

The wealth of ‘big data’ in healthcare is revolutionising our approach to R&D. We are already seeing how the next generation of medicines are being shaped by our ability to capture, interpret and apply data. Combining insight from clinical health records with large-scale genomics data is enabling scientists to better predict disease outcomes in the clinic. However, our team have been investigating connecting multiple data stacks from anywhere and of any type, from proprietary data to real world evidence, even social media platforms. This data stack would truly allow us to map 360 degree views of patient journeys and gain understanding of the interplay between ‘nature’ (from genetic information) and ‘nurture’ (environmental data e.g. smartphone/ sensor data) to make breakthroughs in science and ultimately patient care. In 2016, the team hopes to begin a collaboration to create such a complex data stacks in the oncology therapy area, aiming to get targeted medicines to genetically matched patients faster.

“A key to advancing drug delivery in the next decade for RNA therapeutics will be to enhance trafficking and cellular uptake, to and by the desired tissue and cell type.” Malin Lemurell, Targeted Drug Delivery Futures Lead Introduction

Delivering the next wave of life-changing medicines requires a new way of thinking. To ensure the IMED Biotech Unit remains at the cutting-edge of scientific innovation, we established our IMED Futures programme.

Below Worldwide transportation systems, communication networks and energy infrastructures

CGI images of AstraZeneca’s new Global R&D Centre and Corporate Headquarters, Cambridge, UK

110 ©AstraZeneca 2016

astrazeneca.com