hereditary angioedema prophylaxis with plasma kallikrein

1
Figure 2: Representation of the HAE virtual patient population capturing patient variability in the propensity for acute attacks represented by frequency and severity 0% 10% 20% 30% 40% 1 2 3 4 5 6 7 More Monthly attack rate 0% 5% 10% 15% 20% 25% 30% Distribution percentage Distribution percentage PKa inhibitor pharmacokinetics via random sampling Variability in acute attack trigger via random sampling 1 2 3 4 5 6 11 10 9 8 7 More Virtual HAE population Fold increase in FXII auto-activation Patient 1 = (Pt 1 , f 1 , S 1 ) Patient 2 = (Pt 2 , f 2 , S 2 ) Patient n = (Pt 1 , f n , S n ) Contact activation system pharmacodynamic model FXII = factor XII; HAE = hereditary angioedema; PKa = plasma kallikrein. *Lanadelumab is approved as prophylaxis to prevent attacks of HAE in patients ≥12 years of age in the United States and for the routine prevention of attacks in patients ≥12 years of age in Canada. 9,10 Lanadelumab also was approved in the European Union, Australia, and Brazil for the routine prevention of recurrent attacks of HAE in patients ≥12 years of age, 11-13 and in Switzerland for the long-term prevention of attacks of HAE in patients ≥12 years of age. 14 References: 1. Banerji A, et al. JAMA 2018;320:2108-2121. 2. Aygören-Pürsün E, et al. N Engl J Med 2018;379:352-362. 3. Kalfus I, et al. Presented at the Annual Scientific Session of the Western Society of Allergy, Asthma and Immunology; January 20–4, 2019; Maui, HI. 4. Hampton SL, et al. J Allergy Clin Immunol 2019;143:AB39. 5. van der Graaf PH, Benson N. Pharma Res 2011;28:1460-1464. 6. Nussberger J, et al. Lancet 1998;351:1693-1697. 7. Suffritti C, et al. Clin Exp Allergy 2014;44:1503-1514. 8. Osterberg L, Blaschke T. N Engl J Med 2005;353:487-497. 9. Shire. Highlights of prescribing information. Takhzyro™ (lanadelumab-flyo) injection, for subcutaneous use. https://www.accessdata.fda.gov/drugsatfda_docs/ label/2018/761090s000lbl.pdf. Accessed January 6, 2020. 10. Shire Pharma Canada. Product monograph including patient medication information. Takhzyro™ (lanadelumab injection). https://www.shirecanada.com/-/media/shire/shireglobal/shirecanada/pdffiles/product%20information/takhzyro-pm-en.pdf. Accessed January 6, 2020. 11. European Medicines Agency. Takhzyro 300 mg solution for injection. Summary of product characteristics. http://ec.europa.eu/health/documents/community-register/2018/20181122143033/anx_143033_en.pdf. Accessed January 6, 2020. 12. Takhzyro Australian product information. https://www.tga.gov.au/prescription-medicines-registration-new-chemical-entities-australia. Accessed January 6, 2020. 13. Takhzyro Brazilian product information. http://portal.anvisa.gov.br/resultado-de-busca?p_p_id=101&p_p_lifecycle=0&p_p_state=maximized&p_p_mode=view&p_p_col_ id=column 1&p_p_col_count=1&_101_struts_action=%2Fasset_publisher%2Fview_content&_101_assetEntryId=5676578&_101_type=content&_101_groupId=219201&_101_urlTitle =takhzyro-novo-registro&redirect=http%3A%2F%2Fportal.anvisa.gov.br%2Fresultado-de-busca%3Fp_p_id%3D3%26p_p_lifecycle%3D0%26p_p_state%3Dnormal%26p_p_ mode%3Dview%26p_p_col_id%3Dcolumn-1%26p_p_col_count%3D1%26_3_groupId%3D0%26_3_keywords%3Dlanadelumab%26_3_cur%3D1%26_3_struts_action %3D%252Fsearch%252Fsearch%26_3_format%3D%26_3_formDate%3D1441824476958. Accessed January 6, 2020. 14. Swissmedic. Takhzyro, solution for injection. https://www. swissmedic.ch/swissmedic/en/home/humanarzneimittel/authorisations/authorised-medicinal-products-with-new-active-substances/takhzyro_injektionsloesung_lanadelumabum.html. Accessed January 6, 2020. Funding and acknowledgments: We thank all patients, investigators, and their study staff. This study was sponsored by Shire Human Genetic Therapies, Inc., a Takeda company. Under the direction of the authors, Hoa Q. Nguyen, employee of Takeda, provided writing assistance for this poster. Editorial assistance in formatting, proofreading, copyediting, and fact-checking also was provided by Excel Medical Affairs. Shire Human Genetic Therapies, Inc., a Takeda company, provided funding to Excel Medical Affairs for support in writing and editing this poster. The interpretation of the data was made by the authors independently. Rationale Rationale Hereditary angioedema (HAE) is a rare inherited disorder characterized by recurrent episodes (also called attacks) of severe swelling of the skin and mucous membranes. Unopposed contact system activation leads to excess plasma kallikrein (PKa) activity and, subsequently, increased bradykinin, a key mediator of HAE attacks. PKa inhibitors reduce contact system activation and have been approved or are in development for HAE. Lanadelumab, a fully human monoclonal antibody inhibitor of PKa (half-life of ~14 days) approved for HAE prophylaxis in several regions,* reduced attack rates at 300 mg administered every 2 weeks (q2wks) for 26 weeks by ~87% compared with placebo in the phase 3 HELP Study (NCT02586805). 1 We sought to understand the impact of target binding kinetics, pharmacokinetics, and treatment adherence on maintaining HAE disease control with lanadelumab versus small molecule PKa inhibitors (half-life of 20 hours) in development for HAE prophylaxis via a quantitative systems pharmacology (QSP) approach. 2-5 Methods Methods A mechanistic biological model of HAE was developed using QSP that incorporated critical components of the contact system and a virtual HAE patient population (Figures 1 and 2). Cleaved high molecular weight kininogen pharmacodynamic biomarker data and clinical outcomes from lanadelumab clinical studies were used to verify the model within an acceptable range (Figures 3–5). The verified model was used to evaluate the impact of various drug-related factors on HAE disease outcome, including binding potency, treatment adherence (assessed via virtual patient populations by QSP modeling), and half-life (Figures 6–9). Results Results Model development 3 ways to access the poster Mobile and environmentally friendly e-posters No personal information is stored - standard charges may apply. Send an SMS: Text 474396 To 43704 (USA only) +44 7860 033 046 (rest of world) Access online www.shirecongressposters.com/474396 Scan QR code Presented at the American Academy of Asthma, Allergy & Immunology Annual Meeting, March 13–16, 2020, Philadelphia, PA Hereditary Angioedema Prophylaxis With Plasma Kallikrein Inhibitors: Role of Target Hereditary Angioedema Prophylaxis With Plasma Kallikrein Inhibitors: Role of Target Binding Kinetics, Pharmacokinetics, and Treatment Adherence Binding Kinetics, Pharmacokinetics, and Treatment Adherence Rangaraj Narayanan, 1 * Haobin Luo, 2 Zhiwei Zhang, 2 Paul Jasper, 2 Peng Lu, 1 * Salomé Juethner, 1 Dan Sexton 1 1 Shire, a Takeda company, Cambridge, MA, USA; 2 RES Group Inc., Needham, MA, USA *Affiliation at the time the analysis was conducted 327 Conclusions Conclusions A QSP model of excess contact system activation coupled with virtual population of patients with HAE was developed to quantitatively describe the dynamic interactions of the contact factors and PKa inhibitors. The model successfully captures the clinical outcomes of lanadelumab in virtual patients with HAE. The model was applied to investigate the role of target binding and pharmacokinetics and showed that drugs with long half-lives (>2 weeks) had ≥2-fold improved efficacy versus those with short half-lives (20 hours). For small molecules with short half-lives, the model suggested that the more missed doses, the higher the HAE attack occurrence. Drug development programs address noncompliance and nonpersistence via the development of drugs that require less frequent dosing and demonstrate longer half-lives. Model qualification Model application Role of target binding kinetics and pharmacokinetics Impact of treatment nonadherence in patients Figure 6: Monthly attack rates for 150 mg QD oral small molecule PKa inhibitors with different Ki values Ki = 0.30 nM Ki = 0.50 nM 0 1 2 3 4 5 Week –4 Week 0 Week +4 Week +8 Week +12 Week +16 Week +20 Week +24 Week +28 Week +32 Week +36 Attacks/month First dose Last dose Ki = inhibition constant; PKa = plasma kallikrein; QD = once-daily. In the absence of plasma protein binding, for a 150 mg QD dose, the binding affinity needs to be strong enough (eg, <0.3 nM) to effectively suppress attacks. Figure 8: Example pharmacokinetics of drug X 150 mg QD and bradykinin profiles with a nonadherence rate of 20% 0.00 0.02 0.04 0.06 0.08 0.10 0.12 0.14 1 10 100 1000 0 4 8 12 16 20 24 28 Drug X plasma concentration (ng/mL) Week Drug X Bradykinin Bradykinin (nM) HAE = hereditary angioedema; QD = once daily. Nonadherence for daily dosing of drug X could negatively impact the suppression of HAE attacks because each missed dose would reduce drug coverage and make the patient more prone to HAE attacks. Figure 7: Efficacy of drugs with shorter half-lives (20 hours) 0 20 40 60 80 100 120 0 2 4 6 8 10 12 14 Day 95th percentile Mean 5th percentile 0 20 40 60 80 100 –4 0 4 8 12 16 20 24 28 %HKa Week 95th percentile Mean 5th percentile 150 mg QD 0 1 2 3 4 5 6 Week –4 Week 0 Week +4 Week +8 Week +12 Week +16 Week +20 Week +24 Week +28 Week +32 Week +36 Attacks/month Plasma concentration Placebo 110 mg 150 mg First dose Last dose Predicted %HKa Pharmacokinetics of example drug with half-life 20 hours 150 mg QD Hka = cleaved high molecular weight kininogen; QD = once daily; q2wks = every 2 weeks. Figure 1: QSP model of in vivo contact system activation Depot (PKa inhibitor) FXII FXIIa Kallikrein HK + C1-INH B1R/B2R Bradykinin R P P G F S P F R Feedback loop F, k a Central (CL, V c ) Clinical outcome (reduction of HAE attack rate) Prekallikrein Auto-activation C1-INH HKa (key biomarker) C1-INH = C1-inhibitor; CL = clearance; F = fraction of dose absorbed; FXII = factor XII; FXIIa = activated factor XII; HAE = hereditary angioedema; HK = high molecular weight kininogen; HKa = cleaved high molecular weight kininogen; ka = absorption rate constant; PKa = plasma kallikrein; QSP = quantitative systems pharmacology; Vc = central volume of distribution. Figure 3: QSP model approximated observed %PKa inhibition data from phase 1a (NCT01923207) and phase 1b (NCT02093923) lanadelumab studies 0 20 40 60 80 100 0 20 40 60 80 100 120 140 Data Basal prekallikrein (250 nM) Basal prekallikrein (650 nM) 0 20 40 60 80 100 0 20 40 60 80 100 120 140 Data Basal prekallikrein (250 nM) Basal prekallikrein (650 nM) –20 –10 0 10 20 30 40 50 60 70 0 14 28 42 56 70 84 98 112 Dosed Vehicle Basal prekallikrein (250 nM) Basal prekallikrein (650 nM) –20 –10 0 10 20 30 40 50 0 14 28 42 56 70 84 98 112 Time (days) Dosed Vehicle Basal prekallikrein (250 nM) Basal prekallikrein (650 nM) % inhibition of PKa activity % inhibition of PKa activity % inhibition of PKa activity % inhibition of PKa activity 3 mg/kg 1 mg/kg 300 mg 400 mg Time (days) Time (days) Time (days) Phase 1a study: healthy volunteers (single dose) Phase 1b study: patients with HAE (repeated dose) HAE = hereditary angioedema; PKa = plasma kallikrein; QSP = quantitative systems pharmacology. Figure 4: QSP model approximated observed %HKa levels from the phase 3 lanadelumab study (NCT02586805) 0 20 40 60 80 100 –8 –4 0 4 8 12 16 20 24 28 32 36 40 44 %HKa Time (weeks) %HKa for 300 mg q2wks 5th percentile 95th percentile Median Mean HKa = cleaved high molecular weight kininogen; q2wks = every 2 weeks; QSP = quantitative systems pharmacology. Median %HKa (57%) modeled in 1000 virtual patients approximately matched previous reports.6 Figure 5: QSP model prediction of monthly HAE attack rates in 1000 virtual patients approximated observed attack rates from the phase 3 lanadelumab study (NCT02586805) 0 1 2 3 4 5 Week –4 Week 0 Week +4 Week +8 Week +12 Week +16 Week +20 Week +24 Week +28 Attacks/month 150 mg q4wks Data Model 0 1 2 3 4 5 Attacks/month 300 mg q4wks 0 1 2 3 4 5 Attacks/month 300 mg q2wks First dose Last dose Week –4 Week 0 Week +4 Week +8 Week +12 Week +16 Week +20 Week +24 Week +28 Week –4 Week 0 Week +4 Week +8 Week +12 Week +16 Week +20 Week +24 Week +28 HAE = hereditary angioedema; q2wks = every 2 weeks; q4wks = every 4 weeks; QSP = quantitative systems pharmacology. Model suggests that drugs with shorter half-lives and weaker binding affinity have higher HKa and attack rates (≥2-fold greater) compared with lanadelumab (half-life of ~14 days) at 300 mg q2wks dosing. Impact of treatment nonadherence in patients Pharmacology adherence rates to daily oral therapy in the real-world setting report an 80% mean adherence, with a range of ~65–90%. 8 Nonadherence was studied for drugs with shorter half-lives (drug X) using population simulations with an assumed average of occurrence of nonadherences (random missing dose) of 20% for a virtual patient population (ie, 6 missed doses in a 30-day month). Figure 9: Simulated numbers of monthly attacks during treatment of drug X 150 mg QD with different nonadherence rates 0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 Full adherence 15% missed dose 20% missed dose 30% missed dose 40% missed dose 50% missed dose Attacks/month Placebo Drug X 150 mg QD HAE = hereditary angioedema; QD = once daily. The monthly attack profiles showed that the more missed doses, the higher the HAE attack occurrence. Simulations showed 6–8 days of randomly missed doses out of 4 weeks daily dosing could translate to a ~2-fold efficacy decline. At ≥50% missed dose scenario, treatment with drug results in marginal efficacy. %reduction of attack rates compared with placebo Full adherence 15% missed dose 20% missed dose 30% missed dose 40% missed dose 50% missed dose Drug X 150 mg QD 53.9 41.9 35.5 23.9 21.3 13.2 Frequency (f): number of attack events for a patient was randomly simulated from Poison distribution, with an average of 2.2 attacks per month. Severity (S): severity of attack was modeled as an increase in FXII auto-activation. The increase of FXII auto-activation (assumed normal distribution) was sampled so that the bradykinin levels during attack are in line with literature range. 6 Simulated HAE attacks occurred when bradykinin levels exceeded a predefined threshold plasma concentration in patients with HAE (20 pM). 7

Upload: others

Post on 03-Jan-2022

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Hereditary Angioedema Prophylaxis With Plasma Kallikrein

Figure 2: Representation of the HAE virtual patient population capturing patient variability in the propensity for acute attacks represented by frequency and severity

0%

10%

20%

30%

40%

1 2 3 4 5 6 7 MoreMonthly attack rate

0%5%

10%15%20%25%30%

Dis

trib

utio

n pe

rcen

tage

Dis

trib

utio

n pe

rcen

tage

PKa inhibitor pharmacokineticsvia random sampling

Variability in acute attack triggervia random sampling

1 2 3 4 5 6 1110987 More

Virtual HAE population

Fold increase in FXII auto-activation

Patient1 = (Pt1, f1, S1)Patient2 = (Pt2, f2, S2)Patientn = (Pt1, fn, Sn)

Contact activation system pharmacodynamic model

FXII = factor XII; HAE = hereditary angioedema; PKa = plasma kallikrein.

*Lanadelumab is approved as prophylaxis to prevent attacks of HAE in patients ≥12 years of age in the United States and for the routine prevention of attacks in patients ≥12 years of age in Canada.9,10 Lanadelumab also was approved in the European Union, Australia, and Brazil for the routine prevention of recurrent attacks of HAE in patients ≥12 years of age,11-13 and in Switzerland for the long-term prevention of attacks of HAE in patients ≥12 years of age.14

References: 1. Banerji A, et al. JAMA 2018;320:2108-2121. 2. Aygören-Pürsün E, et al. N Engl J Med 2018;379:352-362. 3. Kalfus I, et al. Presented at the Annual Scientific Session of the Western Society of Allergy, Asthma and Immunology; January 20–4, 2019; Maui, HI. 4. Hampton SL, et al. J Allergy Clin Immunol 2019;143:AB39. 5. van der Graaf PH, Benson N. Pharma Res 2011;28:1460-1464. 6. Nussberger J, et al. Lancet 1998;351:1693-1697. 7. Suffritti C, et al. Clin Exp Allergy 2014;44:1503-1514. 8. Osterberg L, Blaschke T. N Engl J Med 2005;353:487-497. 9. Shire. Highlights of prescribing information. Takhzyro™ (lanadelumab-flyo) injection, for subcutaneous use. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/761090s000lbl.pdf. Accessed January 6, 2020. 10. Shire Pharma Canada. Product monograph including patient medication information. Takhzyro™ (lanadelumab injection). https://www.shirecanada.com/-/media/shire/shireglobal/shirecanada/pdffiles/product%20information/takhzyro-pm-en.pdf. Accessed January 6, 2020. 11. European Medicines Agency. Takhzyro 300 mg solution for injection. Summary of product characteristics. http://ec.europa.eu/health/documents/community-register/2018/20181122143033/anx_143033_en.pdf. Accessed January 6, 2020. 12. Takhzyro Australian product information. https://www.tga.gov.au/prescription-medicines-registration-new-chemical-entities-australia. Accessed January 6, 2020. 13. Takhzyro Brazilian product information. http://portal.anvisa.gov.br/resultado-de-busca?p_p_id=101&p_p_lifecycle=0&p_p_state=maximized&p_p_mode=view&p_p_col_id=column 1&p_p_col_count=1&_101_struts_action=%2Fasset_publisher%2Fview_content&_101_assetEntryId=5676578&_101_type=content&_101_groupId=219201&_101_urlTitle =takhzyro-novo-registro&redirect=http%3A%2F%2Fportal.anvisa.gov.br%2Fresultado-de-busca%3Fp_p_id%3D3%26p_p_lifecycle%3D0%26p_p_state%3Dnormal%26p_p_ mode%3Dview%26p_p_col_id%3Dcolumn-1%26p_p_col_count%3D1%26_3_groupId%3D0%26_3_keywords%3Dlanadelumab%26_3_cur%3D1%26_3_struts_action %3D%252Fsearch%252Fsearch%26_3_format%3D%26_3_formDate%3D1441824476958. Accessed January 6, 2020. 14. Swissmedic. Takhzyro, solution for injection. https://www.swissmedic.ch/swissmedic/en/home/humanarzneimittel/authorisations/authorised-medicinal-products-with-new-active-substances/takhzyro_injektionsloesung_lanadelumabum.html. Accessed January 6, 2020.

Funding and acknowledgments: We thank all patients, investigators, and their study staff. This study was sponsored by Shire Human Genetic Therapies, Inc., a Takeda company. Under the direction of the authors, Hoa Q. Nguyen, employee of Takeda, provided writing assistance for this poster. Editorial assistance in formatting, proofreading, copyediting, and fact-checking also was provided by Excel Medical Affairs. Shire Human Genetic Therapies, Inc., a Takeda company, provided funding to Excel Medical Affairs for support in writing and editing this poster. The interpretation of the data was made by the authors independently.

RationaleRationale ■ Hereditary angioedema (HAE) is a rare inherited disorder characterized by recurrent episodes (also called attacks) of severe swelling of the skin and mucous membranes.

■ Unopposed contact system activation leads to excess plasma kallikrein (PKa) activity and, subsequently, increased bradykinin, a key mediator of HAE attacks.■ PKa inhibitors reduce contact system activation and have been approved or are in development for HAE.■ Lanadelumab, a fully human monoclonal antibody inhibitor of PKa (half-life of ~14 days) approved for HAE prophylaxis in several regions,* reduced attack

rates at 300 mg administered every 2 weeks (q2wks) for 26 weeks by ~87% compared with placebo in the phase 3 HELP Study (NCT02586805).1

■ We sought to understand the impact of target binding kinetics, pharmacokinetics, and treatment adherence on maintaining HAE disease control with lanadelumab versus small molecule PKa inhibitors (half-life of 20 hours) in development for HAE prophylaxis via a quantitative systems pharmacology (QSP) approach.2-5

MethodsMethods■ A mechanistic biological model of HAE was developed using QSP that incorporated critical components of the contact system and a virtual HAE patient

population (Figures 1 and 2).■ Cleaved high molecular weight kininogen pharmacodynamic biomarker data and clinical outcomes from lanadelumab clinical studies were used to verify the

model within an acceptable range (Figures 3–5).■ The verified model was used to evaluate the impact of various drug-related factors on HAE disease outcome, including binding potency, treatment

adherence (assessed via virtual patient populations by QSP modeling), and half-life (Figures 6–9).

ResultsResultsModel development

3 ways to access the poster

Mobile and environmentally friendly e-posters

No personal information is stored - standard charges may apply.

Send an SMS:Text 474396

To 43704 (USA only)+44 7860 033 046 (rest of world)

Access online www.shirecongressposters.com/474396

Scan QR code

Presented at the American Academy of Asthma, Allergy & Immunology Annual Meeting, March 13–16, 2020, Philadelphia, PA

Hereditary Angioedema Prophylaxis With Plasma Kallikrein Inhibitors: Role of Target Hereditary Angioedema Prophylaxis With Plasma Kallikrein Inhibitors: Role of Target Binding Kinetics, Pharmacokinetics, and Treatment AdherenceBinding Kinetics, Pharmacokinetics, and Treatment Adherence

Rangaraj Narayanan,1* Haobin Luo,2 Zhiwei Zhang,2 Paul Jasper,2 Peng Lu,1* Salomé Juethner,1 Dan Sexton1

1Shire, a Takeda company, Cambridge, MA, USA; 2RES Group Inc., Needham, MA, USA*Affiliation at the time the analysis was conducted

327

ConclusionsConclusions■ A QSP model of excess contact system activation coupled with virtual population of patients with HAE was developed to quantitatively

describe the dynamic interactions of the contact factors and PKa inhibitors. The model successfully captures the clinical outcomes oflanadelumab in virtual patients with HAE.

■ The model was applied to investigate the role of target binding and pharmacokinetics and showed that drugs with long half-lives (>2 weeks)had ≥2-fold improved efficacy versus those with short half-lives (20 hours).

■ For small molecules with short half-lives, the model suggested that the more missed doses, the higher the HAE attack occurrence.■ Drug development programs address noncompliance and nonpersistence via the development of drugs that require less frequent dosing

and demonstrate longer half-lives.

Model qualification

Model applicationRole of target binding kinetics and pharmacokinetics Impact of treatment nonadherence in patients

Figure 6: Monthly attack rates for 150 mg QD oral small molecule PKa inhibitors with different Ki values

Ki = 0.30 nM

Ki = 0.50 nM

0

1

2

3

4

5

Week–4

Week0

Week+4

Week+8

Week+12

Week+16

Week+20

Week+24

Week+28

Week+32

Week+36

Att

acks

/mo

nth

First dose Last dose

Ki = inhibition constant; PKa = plasma kallikrein; QD = once-daily.

In the absence of plasma protein binding, for a 150 mg QD dose, the binding affinity needs to be strong enough (eg, <0.3 nM) to effectively suppress attacks.

Figure 8: Example pharmacokinetics of drug X 150 mg QD and bradykinin profiles with a nonadherence rate of 20%

0.00

0.02

0.04

0.06

0.08

0.10

0.12

0.14

1

10

100

1000

0 4 8 12 16 20 24 28

Dru

g X

pla

sma

con

cen

trat

ion

(n

g/m

L)

Week

Drug X Bradykinin

Bra

dyk

inin

(n

M)

HAE = hereditary angioedema; QD = once daily.

Nonadherence for daily dosing of drug X could negatively impact the suppression of HAE attacks because each missed dose would reduce drug coverage and make the patient more prone to HAE attacks.

Figure 7: Efficacy of drugs with shorter half-lives (20 hours)

0

20

40

60

80

100

120

0 2 4 6 8 10 12 14Day

95th percentile Mean 5th percentile

0

20

40

60

80

100

–4 0 4 8 12 16 20 24 28

%H

Ka

Week

95th percentile Mean 5th percentile150 mg QD

0

1

2

3

4

5

6

Week–4

Week0

Week+4

Week+8

Week+12

Week+16

Week+20

Week+24

Week+28

Week+32

Week+36

Att

acks

/mo

nth

Plas

ma

con

cen

trat

ion

Placebo 110 mg 150 mg

First dose Last dose

Predicted %HKaPharmacokinetics of example drug with half-life 20 hours

150 mg QD

Hka = cleaved high molecular weight kininogen; QD = once daily; q2wks = every 2 weeks.

Figure 1: QSP model of in vivo contact system activation

Depot(PKa inhibitor)

FXII FXIIa

Kallikrein

HK +

C1-INH

B1R/B2R

BradykininR P P G F S P F R

Feedbackloop

F, ka

Central(CL, Vc)

Clinical outcome(reduction of HAE

attack rate)

Prekallikrein

Auto-activation

C1-INH

HKa(key biomarker)

C1-INH = C1-inhibitor; CL = clearance; F = fraction of dose absorbed; FXII = factor XII; FXIIa = activated factor XII; HAE = hereditary angioedema; HK = high molecular weight kininogen; HKa = cleaved high molecular weight kininogen; ka = absorption rate constant; PKa = plasma kallikrein; QSP = quantitative systems pharmacology; Vc = central volume of distribution.

Figure 3: QSP model approximated observed %PKa inhibition data from phase 1a (NCT01923207) and phase 1b (NCT02093923) lanadelumab studies

0

20

40

60

80

100

0 20 40 60 80 100 120 140

DataBasal prekallikrein (250 nM)Basal prekallikrein (650 nM)

0

20

40

60

80

100

0 20 40 60 80 100 120 140

DataBasal prekallikrein (250 nM)Basal prekallikrein (650 nM)

–20–10

010203040506070

0 14 28 42 56 70 84 98 112

DosedVehicleBasal prekallikrein (250 nM)Basal prekallikrein (650 nM)

–20

–10

0

10

20

30

40

50

0 14 28 42 56 70 84 98 112Time (days)

DosedVehicleBasal prekallikrein (250 nM)Basal prekallikrein (650 nM)

% in

hib

itio

n o

fPK

a ac

tivi

ty%

inh

ibit

ion

of

PKa

acti

vity

% in

hib

itio

n o

fPK

a ac

tivi

ty%

inh

ibit

ion

of

PKa

acti

vity

3 mg/kg 1 mg/kg

300 mg 400 mg

Time (days)

Time (days) Time (days)

Phase 1a study: healthy volunteers (single dose)

Phase 1b study: patients with HAE (repeated dose)

HAE = hereditary angioedema; PKa = plasma kallikrein; QSP = quantitative systems pharmacology.

Figure 4: QSP model approximated observed %HKa levels from the phase 3 lanadelumab study (NCT02586805)

0

20

40

60

80

100

–8 –4 0 4 8 12 16 20 24 28 32 36 40 44

%H

Ka

Time (weeks)

%HKa for 300 mg q2wks5th percentile 95th percentileMedian Mean

HKa = cleaved high molecular weight kininogen; q2wks = every 2 weeks; QSP = quantitative systems pharmacology.

Median %HKa (57%) modeled in 1000 virtual patients approximately matched previous reports.6

Figure 5: QSP model prediction of monthly HAE attack rates in 1000 virtual patients approximated observed attack rates from the phase 3 lanadelumab study (NCT02586805)

012345

Week –4 Week 0 Week +4 Week +8 Week +12 Week +16 Week +20 Week +24 Week +28

Att

acks

/mo

nth

150 mg q4wks Data Model

012345

Att

acks

/mo

nth

300 mg q4wks

012345

Att

acks

/mo

nth

300 mg q2wks

First dose Last dose

Week –4 Week 0 Week +4 Week +8 Week +12 Week +16 Week +20 Week +24 Week +28

Week –4 Week 0 Week +4 Week +8 Week +12 Week +16 Week +20 Week +24 Week +28

HAE = hereditary angioedema; q2wks = every 2 weeks; q4wks = every 4 weeks; QSP = quantitative systems pharmacology.

Model suggests that drugs with shorter half-lives and weaker binding affinity have higher HKa and attack rates (≥2-fold greater) compared with lanadelumab (half-life of ~14 days) at 300 mg q2wks dosing.

Impact of treatment nonadherence in patients■ Pharmacology adherence rates

to daily oral therapy in thereal-world setting report an 80%mean adherence, with a rangeof ~65–90%.8

■ Nonadherence was studied fordrugs with shorter half-lives(drug X) using populationsimulations with an assumedaverage of occurrence ofnonadherences (random missingdose) of 20% for a virtual patientpopulation (ie, 6 missed doses ina 30-day month).

Figure 9: Simulated numbers of monthly attacks during treatment of drug X 150 mg QD with different nonadherence rates

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

Fulladherence

15%missed dose

20%missed dose

30%missed dose

40%missed dose

50%missed dose

Att

acks

/mo

nth

Placebo Drug X 150 mg QD

HAE = hereditary angioedema; QD = once daily.

The monthly attack profiles showed that the more missed doses, the higher the HAE attack occurrence. Simulations showed 6–8 days of randomly missed doses out of 4 weeks daily dosing could translate to a ~2-fold efficacy decline. At ≥50% missed dose scenario, treatment with drug results in marginal efficacy.

%reduction of attack rates compared with placebo

Fulladherence

15%misseddose

20%misseddose

30%misseddose

40%misseddose

50%misseddose

Drug X 150 mg QD

53.9 41.9 35.5 23.9 21.3 13.2

■ Frequency (f): number of attack events fora patient was randomly simulated from Poisondistribution, with an average of 2.2 attacksper month.

■ Severity (S): severity of attack was modeledas an increase in FXII auto-activation. Theincrease of FXII auto-activation (assumednormal distribution) was sampled so that thebradykinin levels during attack are in line withliterature range.6

■ Simulated HAE attacks occurred whenbradykinin levels exceeded a predefinedthreshold plasma concentration in patientswith HAE (20 pM).7