Adoption by CHMP for release for consultation 09 July Start of public consultation 15 August 2012

1 2 3 4 5 09 July 2012 EMA/CHMP/450916/2012 Committee for Medicinal Products for Human Use (CHMP) 8 Guideline on clinical investigation of medicina...
1 downloads 0 Views 225KB Size
1 2 3 4 5

09 July 2012 EMA/CHMP/450916/2012 Committee for Medicinal Products for Human Use (CHMP)

8

Guideline on clinical investigation of medicinal products for prevention of stroke and systemic embolic events in patients with non-valvular atrial fibrillation

9

Draft

6 7

10 Draft Agreed by Cardiovascular Working Party 1 Adoption by CHMP for release for consultation Start of public consultation End of consultation (deadline for comments)

28 March 2012 09 July 2012 15 August 2012 15 February 2013

11 12 Comments should be provided using this template. The completed comments form should be sent to [email protected] 13 14 Keywords

Stroke, systemic embolism, atrial fibrillation, guidelines, anticoagulant, CHMP

15

16

17

7 Westferry Circus ● Canary Wharf ● London E14 4HB ● United Kingdom Telephone +44 (0)20 7418 8400 Facsimile +44 (0)20 7418 8613 E-mail [email protected] Website www.ema.europa.eu

An agency of the European Union

© European Medicines Agency, 2012. Reproduction is authorised provided the source is acknowledged.

20

Guideline on clinical investigation of medicinal products for prevention of stroke and systemic embolic events in patients with non-valvular atrial fibrillation

21

Draft

22

Table of contents

23

Executive summary ..................................................................................... 3

24

1. Introduction (background)...................................................................... 3

25

2. Scope....................................................................................................... 3

26

3. Legal basis .............................................................................................. 3

27

4. Assessment of efficacy criteria ................................................................ 4

28 29

4.1. Primary efficacy outcome .................................................................................... 4 4.2. Secondary outcomes .......................................................................................... 4

30

5. Methods to assess efficacy ...................................................................... 5

31 32

5.1. Primary efficacy outcome .................................................................................... 5 5.2. Secondary outcomes .......................................................................................... 5

33

6. Selection of patients................................................................................ 6

34

6.1. Study population................................................................................................ 6

35 36

6.2. Inclusion criteria ................................................................................................ 6 6.3. Exclusion criteria................................................................................................ 6

37

7. Strategy design ....................................................................................... 6

38

8. Safety aspects ......................................................................................... 9

39

9. References ............................................................................................ 14

18 19

40

2/15

41 42

Executive summary

43

Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia, occurring in 1–2% of the

44

general population. AF confers a 5-fold risk of stroke, and one in five of all strokes is attributed to this

45

arrhythmia.

46

Current Note for Guidance on Antiarrhythmics (CPMP/EWP/237/95) and its addendum on atrial

47

fibrillation and flutter (EMA/CHMP/EWP/213056/2010) do not cover stroke prevention. The aim of this

48

guideline is to provide guidance to industry when performing trials to develop drugs in prevention of

49 50

stroke and systemic embolic events (SEE) in patients with AF.

51 52

1. Introduction (background)

53

AF is the most common sustained cardiac arrhythmia, occurring in 1–2% of the general population,

54

with 45% of diagnoses being in patients 75 years and older [1]. Over 6 million Europeans suffer from

55

this arrhythmia, and its prevalence is estimated to at least double in the next 50 years as the

56

population ages [2,3]. Based on the presentation and duration of the arrhythmia, AF is classified as:

57

first diagnosed, paroxysmal, persistent and permanent AF [2]. Ischaemic strokes in association with AF

58

are often fatal, and those patients who survive are left more disabled by their stroke and more likely to

59

suffer a recurrence than patients with other causes of stroke. Current recommendations for

60

antithrombotic therapy are based on the presence (or absence) of risk factors for stroke and

61

thromboembolism [2,4]. The simplest risk assessment scheme in non-valvular AF is the CHADS 2 score

62

[cardiac failure, hypertension, age, diabetes, prior stroke or TIA (transient ischaemic attack) (doubled)]

63

[1]. The original validation of this scheme classified a CHADS 2 score of 0 as low risk, 1–2 as moderate

64

risk, and >2 as high risk. In patients with a CHADS 2 score of ≥2, chronic anticoagulation therapy with

65

a vitamin K antagonist (VKA) is currently recommended in a dose adjusted manner to achieve an

66

International Normalised Ratio (INR) value in the range of 2.0–3.0 [2]. In these patients, antiplatelet

67

therapy could be considered as alternative therapy only when VKA therapy is unsuitable. In patients

68

with a CHADS 2 score of 0–1, or where a more detailed stroke risk assessment is indicated, it is

69

recommended to use a more comprehensive risk factor-based approach (e.g. CHA 2 DS 2 -VASc score)

70

[5].

71

Approximately only 30-60% of eligible patients receive oral anticoagulation with VKA and its use in

72

clinical practice is challenging for several reasons, including a narrow therapeutic window, variability in

73

response, interactions and laboratory standardisation [Ansell et al, 2008]. On average, patients may

74

stay within the therapeutic INR range of 2.0–3.0 for 60–65% of the time in controlled clinical trials, but

75

many ‘real-life’ studies suggest that this figure may be 25 cm2, or > 100 cm2 if there was a traumatic cause; intramuscular hematoma documented by

437

ultrasonography without compartment syndrome; excessive wound hematoma; macroscopic hematuria

438

(spontaneous or lasting >24 h if associated with an intervention); epistaxis or gingival bleeding that

439

requires tamponade or other medical intervention, or bleeding after venipuncture for >5 min;

440

hemoptysis, hematemesis or spontaneous rectal bleeding requiring endoscopy or other medical

441 442 443 444

intervention.

445

Other non-major bleedings include other overt bleeding events that do not meet the criteria for major

Other non-major bleedings

446

bleed or clinically relevant non-major bleed (e.g.: epistaxis that does not require medical attention or

447 448 449 450

change in antithrombotic therapy).

451

The use of the following composite bleeding endpoints is recommended:

452

Composite bleeding endpoints of interest

-

453 454

bleeding and/or a clinically relevant non-major bleeding. -

455 456 457 458 459 460

Clinically relevant bleeding: defined as the rate of patients experiencing at least one major Non-major bleeding: defined as the rate of patients experiencing at least one clinically relevant non-major bleeding or other non-major bleeding.

-

Total bleeding: defined as the rate of patients experiencing at least one major bleeding, clinically relevant non-major bleeding or other non-major bleeding.

Other parameters related to bleed

461

As support for the conclusions drawn from the main safety criteria, other bleedings related parameters

462

are recommended to be recorded during the studies e.g.:

463 464

-

465 466

Laboratory parameters: haemoglobin plasma level, haematocrit and red cell count changes during the treatment period,

-

Bleeding index (mean, ±SD) calculated in each patient as the number of units of packed red

467

cells or whole blood transfused plus the haemoglobin values pre-randomisation minus the

468

haemoglobin values at the end of treatment period.

469

-

470 471

-

472 473 474 475 476 477

Patients with bleeding index ≥ 2 at the end of treatment period relative to haemoglobin prerandomisation levels (n, %). Patients receiving transfusion of packed red cells (n, %) (homologous and autologous transfusions need to be distinguished).

-

Transfusion volume (mL; mean, ±SD) and transfusion units (U; mean, ±SD) during the treatment period (homologous and autologous transfusions need to be distinguished).

Report and collection of bleeding events and related parameters

478

The population included in the assessment of bleeding events should correspond with those subjects

479

who have received at least one dose of the study drug (either active or placebo) (i.e.: the safety

480

population).

481

11/15

482

The period for collection of these data should be identical in all treatment groups, starting at the time

483

of the administration of the first dose of study drug (either active or placebo) in any of the treatment

484

groups, until the antithrombotic effect of study drugs is not detectable, and after study drugs have

485

been cleared from plasma.

486 487

The decrease in the haemoglobin level ≥ 2 g/dL should be considered relative to the pre-randomisation

488

level (usually corresponding with the pre-operative haemoglobin).

489 490

The use of a fecal occult blood test (FOBT) at screening visit and during treatment at regular intervals

491

is encouraged, since long-term antithrombotic therapy may be associated with unperceived

492 493 494

gastrointestinal blood loss.

The need for specific antidote and laboratory monitoring

495 496

The development of a specific antidote for new antithrombotics when given at high doses for long-term,

497

as in stroke prevention in AF, is highly recommended given the potential for life-threatening bleeding

498

events in standard practice. Phase I studies are likely to provide a neutralising dose, but they are not

499

expected to address the complex interplay of physiology, concomitant measures (i.e.: blood

500

transfusions, use of plasma expanders, etc) and potential for increased thrombogenicity after

501

administration of the antidote in patients who experience life-threatening bleed. This can be followed

502

by a proof-of-principle study in a small subset of patients with life-threatening bleeding to demonstrate

503

the efficacy and safety in a heterogeneous population. A post authorisation safety study (PASS) will be

504

needed to provide further data. A randomised clinical study will be difficult to perform taking into

505

account the heterogeneity of the population and differences in standard care between the various

506

centres. Furthermore, the potential comparator is difficult to be established, since, up to date, non-

507

specific procoagulant agents are not licensed for reversal of the new agents and may be associated

508

with an increased risk of thrombosis.

509 510

The development of a test for laboratory monitoring of the anticoagulant effect of new agents is highly

511

recommended as well. Even if the new drugs have no monitoring requirements and monitoring has not

512

been applied in pivotal studies, there are potential situations in standard practice where this

513

information might be useful (e.g.: impaired renal function, clinically relevant drug-drug interactions,

514

overdose, measurement of treatment compliance, etc.) that will recommend having it.

515 516 517

8.2

518

The mechanism of action and pharmacological class of the medicinal product under investigation may

519

suggest specific aspects of safety evaluation (e.g. platelet counts, antibody detection, renal and liver

Other events

520

function parameters, hypercoagulability markers to assess a possible rebound hypercoagulation after

521

treatment cessation, etc.) that should be considered for incorporation into the entire development

522

programme.

523 524

If there is a potential for drug-induced liver injury (DILI) with the study drugs (experimental and/or

525

control), an algorithm for hepatic monitoring has to be included in the protocol [13]. Available

526

regulatory guidance on DILI should be followed [17].

527 528

Special attention should be paid to hypersensitivity reactions of the skin and other organs (especially

529

liver, kidney, lungs), changes in blood cells, and hepatitis.

530

12/15

531

For biotechnology derived product(s), immunogenicity should be evaluated prospectively. The type of

532

antibody (e.g. neutralising) and incidence of immune mediated adverse events should be assessed and

533 534 535 536

clearly documented.

537

This should be assessed as dictated by the product and the target population.

538

In general, the following groups might require specific evaluation:

8.3 Special populations

539

-

elderly

540

-

renal insufficiency (moderate, severe)

541

-

liver disease

542 543

Regarding the elderly, it is important to determine whether or not the pharmacokinetic behaviour,

544

pharmacodynamics, disease-drug, drug-drug interactions and clinical response of the drug in this

545

population is different from that in younger adults. Therefore, to assess the benefit/risk balance of a

546

drug that will be used in the geriatric population, a representative number of patients >65 years and

547

>75 years should be appropriately represented in clinical trials (ICH E7).

548 549

The identification of the more appropriate dose in these special populations, in particular in elderly

550

patients, is a matter of utmost importance. Any dose adaptation in these populations should be

551

appropriately justified.

552 553

As long as there is a reasonable representation of the above sub-groups of patients in the main

554

therapeutic study/es, a separate study is not considered necessary.

555

Safety in special populations should be prospectively assessed for inclusion of the sub-groups in SPC.

13/15

556

9. References

557

1. Gage BF, Waterman AD, Shannon W, et al. Validation of clinical classification schemes for predicting

558

stroke: results from the national registry of atrial fibrillation. JAMA. 2001; 285: 2864-70.

559

2. Camm AJ, Kirchhof P, Lip GY, et al. Guidelines for the management of atrial fibrillation: the Task

560

Force for the Management of Atrial Fibrillation of the European Society of Cardiology (ESC). Eur Heart

561

J. 2010; 31: 2369-429.

562

3. Furie KL, Kasner SE, Adams RJ, et al. Guidelines for the Prevention of Stroke in Patients With Stroke

563

or Transient Ischemic Attack: A Guideline for Healthcare Professionals From the American Heart

564

Association/American Stroke Association. Stroke. 2011; 42: 227-76.

565

4. Stroke in AF working group. Independent predictors of stroke in patients with atrial fibrillation: a

566

systematic review. Neurology. 2007; 69: 546–54.

567

5. Lip GY, Nieuwlaat R, Pisters R, et al. Refining clinical risk stratification for predicting stroke and

568

thromboembolism in atrial fibrillation using a novel risk factor-based approach: The Euro Heart Survey

569

on Atrial Fibrillation. Chest 2010; 137: 263–72.

570

6. Ansell J, Hirsh J, Hylek E, et al. Pharmacology and management of the vitamin K antagonists:

571

American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest.

572

2008; 133(6 Suppl): 160S-198S.

573

7. Lip GY, Andreotti F, Fauchier L, et al. Bleeding risk assessment and management in atrial fibrillation

574

patients: a position document from the European Heart Rhythm Association, endorsed by the European

575

Society of Cardiology Working Group on Thrombosis. Europace. 2011; 13: 723-46.

576

8. Pisters R, Lane DA, Nieuwlaat R, et al. A novel user-friendly score (HAS-BLED) to assess 1-year risk

577

of major bleeding in patients with atrial fibrillation: the Euro Heart Survey. Chest. 2010; 138: 1093-

578

100.

579

9. ROCKET AF Study Investigators. Rivaroxaban-once daily, oral, direct factor Xa inhibition compared

580

with vitamin K antagonism for prevention of stroke and Embolism Trial in Atrial Fibrillation: rationale

581

and design of the ROCKET AF study. Am Heart J. 2010; 159: 340-7.

582

10. Mueck W, Lensing AW, Agnelli G, et al. Rivaroxaban: population pharmacokinetic analyses in

583

patients treated for acute deep-vein thrombosis and exposure simulations in patients with atrial

584

fibrillation treated for stroke prevention. Clin Pharmacokinet. 2011; 50: 675-86.

585

11. Rosendaal F, Cannegieter S, van der Meer F, et al. A method to determine the optimal intensity of

586

oral anticoagulant therapy. Thromb Haemost. 1993; 69: 236-9.

587

12. Connolly SJ, Pogue J, Eikelboom J, et al; ACTIVE W Investigators. Benefit of oral anticoagulant

588

over antiplatelet therapy in atrial fibrillation depends on the quality of international normalized ratio

589

control achieved by centers and countries as measured by time in therapeutic range. Circulation. 2008;

590

118: 2029-37.

591

13. Ezekowitz MD, Connolly S, Parekh A, et al. Rationale and design of RE-LY: randomized evaluation

592

of long-term anticoagulant therapy, warfarin, compared with dabigatran. Am Heart J. 2009; 157: 805-

593

10.

594

14. Schulman S, Kearon C; Subcommittee on Control of Anticoagulation of the Scientific and

595

Standardization Committee of the International Society on Thrombosis and Haemostasis. Definition of

596

major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients. J

597

Thromb Haemost. 2005; 3: 692-4.

14/15

598

15. Yusuf S, Zhao F, Mehta SR, et al. Effects of clopidogrel in addition to aspirin in patients with acute

599

coronary syndromes without ST-segment elevation. N Engl J Med 2001; 345: 494–502.

600

16. Ruff CT, Giugliano RP, Antman EM, et al. Evaluation of the novel factor Xa inhibitor edoxaban

601

compared with warfarin in patients with atrial fibrillation: design and rationale for the Effective

602

aNticoaGulation with factor xA next GEneration in Atrial Fibrillation-Thrombolysis In Myocardial

603

Infarction study 48 (ENGAGE AF-TIMI 48). Am Heart J. 2010; 160: 635-41.

604

17. Food and Drug Administration. Guidance for Industry. Drug-Induced Liver Injury: Premarketing

605

Clinical Evaluation. Available at: http://www.fda.gov

15/15