Roxadustat (FG-4592; ASP1517; AZD9941) in the Treatment of in Patients with Lower Risk Myelodysplastic Syndrome (LR-MDS) and Low Red Cell (RBC) Transfusion Burden (LTB) Open-Label Data

David Henry1, John Glaspy2, Rosemary Harup3, Moshe Mittleman4, Amy Zhou5, Charles Bradley6, Gopal Saha6, Pamela Bartels6, Robert Leong6, Kin-Hung P. Yu6 Disclosures

• Research Grant from FibroGen, Inc., San Francisco, CA

2 Anemia in Myelodysplastic Syndrome (MDS)

• More than 90% of MDS patients present with anemia at the time of diagnosis1 • Around 60% of MDS patients will experience severe anemia ( <8 g/dL) at some point during the course of their disease1 • Limited anemia treatment options • ESA (Part of MDS anemia treatment guideline, although not approved in the U.S.) • Large doses may be necessary, dosing not-standardized • Loss of effect over time • RBC transfusions • Exposes patients to iron overload • Infection risk • Development of transfusion dependency is associated with a shorter patient survival as well as an increased risk of conversion to AML1

1. John M. Bennett. Consensus statement on iron overload in myelodysplastic syndromes, Am. J. Hematol. 83:858–861, 2008 3 Roxadustat: Novel, First-in-Class Treatment for Anemia

Roxadustat is an oral hypoxia-inducible factor (HIF) prolyl hydroxylase inhibitor • 2019 Nobel Prize winning science is the foundation of roxadustat • Increases hemoglobin (Hb) by mimicking the body’s natural response to low oxygen • Studied for treatment of anemia in Stage 3 to 5 CKD patients, both dialysis and non-dialysis • Approved in China: (dialysis 12/2018, non-dialysis 8/2019) and Japan: (dialysis 9/2019)

HIF-PH1 Enzymes EPO Within or Near 2019 Nobel Prize HIF-α Degrades Rapidly Physiological Range NORMAL in Physiology or Medicine OXYGEN Degradation Iron Transport to the “For their discoveries of how cells sense and HIF- Bone Marrow and adapt to oxygen availability.” α Hemoglobin (Hb) HIF-α Synthesis Awarded Jointly to: Iron Absorption William G. Kaelin Jr. LOW OXYGEN HIF-α HIF-α HIF-β (e.g., High Altitude) Harvard University or Hepcidin Levels Gregg L. Semenza ✕ Gene Johns Hopkins University ROXADUSTAT Transcription Peter J. Ratcliffe Francis Crick Institute London HIF-PH Enzymes Roxadustat Stabilizes HIF-α Production

1. Hypoxia-inducible factor prolyl hydroxylase 4 Roxadustat: Novel, First-in-Class Treatment for Anemia

Roxadustat – Oral hypoxia-inducible factor (HIF) prolyl-hydroxylase inhibitor • Promotes erythropoiesis by increasing endogenous EPO, improving Fe regulation, and reducing hepcidin • Roxadustat is differentiated from ESAs: • Regulates erythropoiesis via HIF stabilization (not an analogue) • Increase EPO receptors in the bone marrow • Improved iron metabolism and bioavailability • Effective erythropoiesis at or near physiologic EPO levels • Effective erythropoiesis in the presence of inflammation • Requires less or no IV iron • Orally administered, three times a week

5 Roxadustat Reduces Hepcidin (Phase 3 DD-CKD)

• Change from baseline to Week 24 (ROCKIES, DD-CKD Study)

Hepcidin Roxadustat Epoetin alfa (n=608) (n=625) Mean baseline values (ng/mL) 275.61 269.62 0

-10

-20

-30

ng/mL) -40 (

-50

-60

Adjusted Adjusted in LS changemean hepcidin P<0.001 -70

6 Roxadustat Global Phase 3 Development in CKD Anemia

16 Phase 3 studies​ including 6 pivotal studies for U.S. FDA submission • 3 placebo-controlled studies in non-dialysis dependent • 3 active-controlled vs. epoetin alfa studies in dialysis (NDD) CKD patients, pooled (N=4277, with average dependent (DD) CKD patients, pooled (N=3880, with average exposure of 1.6 years) exposure of 1.7 years)

Roxa Corrected Anemia in ESA Naive CKD Patients Roxa Corrected and Maintained Hb in Dialysis Patients

NDD (N=4277): Mean Hb over time up to Week 52 (g/dL) DD (N=3857): mean Hb (g/dL) over time

12 Hb change to Week 28–52: 1.85 (Roxa) vs 0.13 (Placebo) P<0.001 11.4 ∆Hb = 1.22 (Roxa) vs 0.99 (Epoetin alfa) P<0.001 11.2 11.5 11

SE 11 SE

± 10.8 ± 10.5 10.6 10 10.4

9.5 10.2 10

MeanHb(g/dL) 9 MeanHb(g/dL) 9.8 8.5 Roxadustat Placebo Baseline Hb = 9.1 g/dL 9.6 Roxadustat Epoetin alfa 8 9.4 00 22 44 66 88 10101212 1414 1616 1818 2020 222424 262828 303232 34 3636 38 4040 42 4444 46 48 50 52 0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30 32 34 36 38 40 42 44 46 48 50 52 Weeks Since Treatment Weeks Since Treatment Group/n Roxadustat 1940 1864 1805 1836 1788 1747 1744 1712 1684 1667 1661 1615 1579 1545 1479 1442 1382 1336 1288 Active Control 1940 1841 1836 1818 1808 1799 1779 1770 1756 1742 1707 1673 1660 1619 1570 1533 1490 1429 1382 7 Efficacious Regardless of Inflammation and Requires Less IV Iron – Dialysis Dependent CKD Patients

• In patients with inflammation (CRP >ULN) and without inflammation (CRP ≤ULN) roxadustat achieved higher Hb increase compared to epoetin alfa • Less IV iron was required in roxadustat patients than in patients on epoetin alfa

DD: Hb (g/dL) Change from Baseline DD: Less Monthly IV Iron Use in Roxadustat to Weeks 28-52 Patients than in Epoetin Alfa Patients 120 Baseline CRP > ULN Baseline CRP ≤ ULN 100 1.4 1.29 1.4 1.27 1.2 1.2 1.05 80 0.96 1 1 60 0.8 0.8 0.6 0.6 40 0.4 0.4 20

0.2 0.2 MeanMonthly IV Use Iron (mg) Roxadustat (N=1929) Epoetin alfa (N=1928) 0 0 0 Roxadustat EPO Roxadustat EPO 1–8 9–16 17–24 25–32 33–40 41–48 49–52 N=723 N=722 N=889 N=912 n Week P<0.0001 P<0.0001 Roxadustat 1929 1847 1761 1681 1590 1499 1406 Epoetin alfa 1928 1885 1823 1736 1659 1582 1485

8 CV Safety of Roxadustat Demonstrated in CKD Patients in MACE, MACE+, All Cause Mortality Safety Endpoints in CKD Patients

• In Nondialysis patients (N=4270): Risks of *MACE, **MACE+, or all-cause mortality in roxadustat patients were comparable to placebo • In Dialysis Dependent Patients (N=3880) • Risk of MACE and all-cause mortality in roxadustat patients were not increased compared to epoetin alfa in DD patients • Roxadustat patients had a lower risk of MACE+ than epoetin alfa patients • In Incident Dialysis Patients (N=1527, subset of DD) • Roxadustat had 30% lower risk of MACE and a 34% lower risk of MACE+ than epoetin alfa • Roxadustat trends toward lower all-cause cause mortality risk relative to epoetin alfa in incident dialysis patients

All-Cause Proportion of Patients Without MACE+ Over Time Hazard Ratio (95% CI)* *MACE **MACE+ Mortality Incident Dialysis Patients, N=1527 1 Non-Dialysis-Dependent (NDD),

N=4270 1.08 (0.94, 1.24) 1.04 (0.91, 1.18) 1.06 (0.91, 1.23) MACE+ (Placebo Comparator, ITT-a)

Without 0.9 Dialysis-Dependent, N=3880 0.86 (0.74, 0.98) 0.96 (0.82, 1.13) 0.96 (0.79, 1.17) (Epoetin Alfa Comparator) (UB<1.0, p=0.028)

P=0.005 Incident Dialysis, N=1527 (subgroup of Dialysis 0.70 (0.51, 0.96) 0.66 (0.50, 0.89) Roxadustat 0.76 (0.52, 1.11) Dependent (UB<1.0, p=0.029) (UB<1.0, p=0.005) (Epoetin Alfa Comparator) Epoetin alfa Proportion of of PatientsProportion 0.7 * MACE = Major adverse cardiovascular event: death, myocardial infarction, and stroke. Time (Months) 3 6 9 12 18 24 30 36 42 48 54 ** MACE+ = MACE plus unstable angina hospitalization, congestive heart failure requiring hospitalization. MACE & MACE+ events were centrally adjudicated by experts blinded to treatment assignments. Epoetin alfa 699 605 511 401 316 246 171 111 50 24 0 Roxadustat 673 594 495 389 304 223 137 87 43 16 0 9 FGCL-4592-082: Phase 3 Study Design

Key Eligibility Criteria Key Endpoints • Adult lower risk primary MDS patients (IPSS- • Primary: Proportion of patients achieved R ≤4.5) with <5% BM blast transfusion independence (TI) for • No del(5q) cytogenic abnormality ≥8 weeks • Low transfusion burden, requiring 1-4 pRBC • Secondary: Proportion of patients achieved transfusion per 8-weeks at baseline a ≥50% reduction in RBC transfusion over any 8 week period • No ESA within 8 weeks of Day 1 • Proportion of patients achieved TI for ≥20 • EPO ≤400 mIU/mL consecutive weeks • Hb ≤10.0 g/dL • Safety: Review of adverse events, serious adverse events

Best Supportive Care Including RBC Transfusion (per institutional criteria) is Permitted During the Study

10 FGCL-4592-082: Study Overview

Two-Part Study: I. Lead-in, open-label, dose finding segment; 3 sequential starting dose cohorts, 8 patients per cohort (N=24) • Primary goal identify starting dose for DB and evaluate safety II. Randomized, double-blind, placebo controlled segment (N= 156, 3:2 randomization) • Primary objectives: evaluate efficacy (transfusion independence) and safety

Open-Label Lead-In

Screening Period Treatment Period (28 days) (52 weeks)

• Lower Risk MDS Patients TIW • Dose adjustment allowed every 8 weeks • Transfusion Dependent • 1.5 mg/kg • Best Supportive Care allowed per protocol (1-4 pRBC last 8 weeks) • 2.0 mg/kg Hb ≤ 10.0g/dL • 2.5 mg/kg

Recording/review of transfusion history 8-16 weeks prior to Day 1 11 FGCL-4592-082: Baseline Characteristics (Open-Label)

N=24 Age, Median (range), Years 73.0 (57-86)

pRBC Transfusion Burden, Median (range) Units/8-Weeks 4.0 (1-6) MDS Duration, Median (range), Years 3.60 (0.1-15.1) IPSS-R Risk Category, Low, n (%) 20 (83.3) IPSS-R Risk Category, Intermediate, n (%) 4 (16.7) IPSS-R Risk Score, Median, (Range) 2.50 (2-3.5) Bone Marrow Blast, Median (%) 2 (0-4) Hemoglobin, Pre-Transfusion, Median (range) g/dL 8.4 (6.7-10.2)

EPO, Median (range), mIU/mL 124.5 (26-468) Ferritin, Median (range) (ng/mL) 1245 (349-1500) TSAT, Median (range), (%) 54 (11-83) Hs-CRP ≤ULN, n (%) 18 (75.0) Hs-CRP >ULN, n (%) 6 (25.0)

12 FGCL-4592-082: Efficacy Assessment (Open-Label)

Weeks 1-28 Efficacy Endpoints Weeks 1-52 (Primary)

Transfusion Independence ≥8 Weeks, n (%) 9 (38%) 10 (42%)

Weeks 1-28 Weeks 1-52

≥50% Reduction in pRBC Over Any 8 Weeks, n (%) 13 (54%) 14 (58%)

• Median (range) number of days without transfusion: 79 (56-361) days • No patient required IV iron • 78% (7 of 9) patients were on 2.5 mg/kg dose at the time of transfusion independence

13 FGCL-4592-082: Adverse Events (Open-Label)

Summary of AEs Commonly Reported AEs (>1 patient)

N=24, n (%) N=24, n (%) Diarrhea 5 (20.8) Subject with Any AE 19 (79.2) Dyspnea 5 (20.8) Possibly Related (PI assessed) 6 (25.0) Bronchitis 4 (16.7) AEs with CTCAE Grade 3 or Higher 5 (20.8) Nausea 4 (16.7) Contusion 3 (12.5) Serious Adverse Events 5 (20.8) Hyperglycemia 3 (12.5) AEs Leading to Discontinuation of Study Treatment 1 (4.2) Peripheral Edema 3 (12.5) AEs Leading to Death 0 Palpitations 3 (12.5) Abdominal Pain 2 (8.3) Anemia 2 (8.3) Arthralgia 2 (8.3) No Conversion to AML AST Increased 2 (8.3) Dizziness 2 (8.3) Epistaxis 2 (8.3) Headache 2 (8.3) Insomnia 2 (8.3) Non-Cardiac Chest Pain 2 (8.3) Pleural Effusion 2 (8.3) Productive Cough 2 (8.3) Upper Respiratory Tract Infection 2 (8.3) 14 FGCL-4592-082: Summary/Conclusion

✔ Roxadustat is a novel, first-in-class HIF stabilizer for treatment for CKD anemia. ✔ In lower-risk, low transfusion burden MDS patients, treatment with roxadustat resulted in 8-week transfusion independence rate of 38% in the first 28 weeks; 54% of patients had ≥50% reduction in pRBC transfusions over any 8 weeks, from baseline. ✔ Roxadustat was generally well tolerated in each dose cohort. ✔ 2.5 mg/kg dose level was selected as starting dose for DB phase because 78% (7 of 9) patients were on this dose at the time of TI in open label phase, with no dose-limiting toxicity encountered. ✔ Roxadustat is a potential new therapy for treatment of anemia in lower-risk, transfusion dependent MDS patients (roxadustat is not approved to treat MDS in any country).

15 Acknowledgements

We thank all the patients, their families, and investigators who participated in the study.

And, many other contributors who made Roxadustat clinical studies possible.

16 Thank You

17