[Skip to Content]
[Skip to Content Landing]

Effect of Therapeutic Drug Monitoring vs Standard Therapy During Infliximab Induction on Disease Remission in Patients With Chronic Immune-Mediated Inflammatory DiseasesA Randomized Clinical Trial

Educational Objective
To learn the effect of therapeutic drug monitoring (TDM) during infliximab induction on clinical outcomes in patients with chronic immune-mediated inflammatory diseases.
1 Credit CME
Key Points

Question  Among patients with chronic immune-mediated inflammatory diseases initiating treatment with infliximab, does proactive therapeutic drug monitoring (TDM) improve clinical remission rates compared with standard therapy?

Findings  In this randomized clinical trial that included 411 patients, the proportion of patients who experienced disease remission after 30 weeks was 50.5% in the TDM group and 53.0% in the standard therapy group, a difference that was not statistically significant.

Meaning  These findings do not support routine use of proactive TDM during infliximab induction for improving disease remission rates.

Abstract

Importance  Proactive therapeutic drug monitoring (TDM), defined as individualized drug dosing based on scheduled monitoring of serum drug levels, has been proposed as an alternative to standard therapy to maximize efficacy and safety of infliximab and other biological drugs. However, whether proactive TDM improves clinical outcomes when implemented at the time of drug initiation, compared with standard therapy, remains unclear.

Objective  To assess whether TDM during initiation of infliximab therapy improves treatment efficacy compared with standard infliximab therapy without TDM.

Design, Setting, and Participants  Randomized, parallel-group, open-label clinical trial of 411 adults with rheumatoid arthritis, spondyloarthritis, psoriatic arthritis, ulcerative colitis, Crohn disease, or psoriasis initiating infliximab therapy in 21 hospitals in Norway. Patients were recruited from March 1, 2017, to January 10, 2019. Final follow-up occurred on November 5, 2019.

Interventions  Patients were randomized 1:1 to receive proactive TDM with dose and interval adjustments based on scheduled monitoring of serum drug levels and antidrug antibodies (TDM group; n = 207) or standard infliximab therapy without drug and antibody level monitoring (standard therapy group; n = 204).

Main Outcomes and Measures  The primary end point was clinical remission at week 30.

Results  Among 411 randomized patients (mean age, 44.7 [SD, 14.9] years; 209 women [51%]), 398 (198 in the TDM group and 200 in the standard therapy group) received their randomized intervention and were included in the full analysis set. Clinical remission at week 30 was achieved in 100 (50.5%) of 198 and 106 (53.0%) of 200 patients in the TDM and standard therapy groups, respectively (adjusted difference, 1.5%; 95% CI, −8.2% to 11.1%; P = .78). Adverse events were reported in 135 patients (68%) and 139 patients (70%) in the TDM and standard therapy groups, respectively.

Conclusions and Relevance  Among patients with immune-mediated inflammatory diseases initiating treatment with infliximab, proactive therapeutic drug monitoring, compared with standard therapy, did not significantly improve clinical remission rates over 30 weeks. These findings do not support routine use of therapeutic drug monitoring during infliximab induction for improving disease remission rates.

Trial Registration  ClinicalTrials.gov Identifier: NCT03074656

Sign in to take quiz and track your certificates

Buy This Activity

JN Learning™ is the home for CME and MOC from the JAMA Network. Search by specialty or US state and earn AMA PRA Category 1 Credit(s)™ from articles, audio, Clinical Challenges and more. Learn more about CME/MOC

CME Disclosure Statement: Unless noted, all individuals in control of content reported no relevant financial relationships. If applicable, all relevant financial relationships have been mitigated.

Article Information

Corresponding Author: Silje Watterdal Syversen, MD, PhD, Division of Rheumatology and Research, Diakonhjemmet Hospital, PO Box 23 Vinderen, N-0319 Oslo, Norway (s.w.syversen@gmail.com).

Accepted for Publication: March 4, 2021.

Author Contributions: Drs Syversen and Sexton had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Drs Syversen, Goll, and Jørgensen contributed equally and are co–first authors. Drs Jahnsen, Bolstad, and Haavardsholm contributed equally and are co–last authors.

Concept and design: Syversen, Goll, Jørgensen, Olsen, Torp, Mørk, Kvien, Jahnsen, Bolstad, Haavardsholm.

Acquisition, analysis, or interpretation of data: Syversen, Goll, Sandanger, Sexton, Olsen, Gehin, Warren, Brun, Klaasen, Karlsen, Noraberg, Zettel, Ljoså, Haugen, Njålla, Bruun, Seeberg, Michelsen, Strand, Skorpe, Blomgren, Bragnes, Dotterud, Thune, Ystrøm, Torp, Mielnik, Kvien, Jahnsen, Bolstad, Haavardsholm.

Drafting of the manuscript: Syversen, Goll, Jørgensen, Sexton, Warren, Blomgren, Torp, Kvien, Bolstad.

Critical revision of the manuscript for important intellectual content: Syversen, Goll, Jørgensen, Sandanger, Sexton, Olsen, Gehin, Warren, Brun, Klaasen, Karlsen, Noraberg, Zettel, Ljoså, Haugen, Njålla, Bruun, Seeberg, Michelsen, Strand, Skorpe, Bragnes, Dotterud, Thune, Ystrøm, Torp, Mielnik, Mørk, Kvien, Jahnsen, Bolstad, Haavardsholm.

Statistical analysis: Syversen, Sexton, Olsen, Brun, Torp, Kvien, Haavardsholm.

Obtained funding: Syversen, Goll, Jørgensen, Torp, Kvien, Bolstad, Haavardsholm.

Administrative, technical, or material support: Syversen, Goll, Sandanger, Sexton, Gehin, Warren, Brun, Klaasen, Noraberg, Zettel, Ljoså, Haugen, Njålla, Bruun, Seeberg, Michelsen, Strand, Skorpe, Bragnes, Dotterud, Thune, Torp, Jahnsen, Bolstad, Haavardsholm.

Supervision: Syversen, Goll, Jørgensen, Michelsen, Ystrøm, Torp, Mørk, Kvien, Jahnsen, Haavardsholm.

Conflict of Interest Disclosures: Dr Syversen reported receipt of personal fees from Thermo Fisher. Dr Goll reported receipt of personal fees from Pfizer, AbbVie, Boehringer Ingelheim, Roche, Orion Pharma, Sandoz, and Novartis. Dr Jørgensen reported receipt of personal fees from Celltrion, AOP Orphan Pharmaceuticals, and Norgine. Dr Gehin reported receipt of personal fees from Roche. Dr Michelsen reported receipt of personal fees from Novartis and grants from Novartis (paid to employer). Dr Dotterud reported receipt of personal fees from LEO Pharma. Dr Mørk reported receipt of personal fees from Novartis Norge, LEO Pharma, ACO Hud Norge, Celgene, AbbVie, Galderma Nordic, and UCB. Dr Kvien reported receipt of grants from AbbVie, Bristol-Myers Squibb, Merck Sharp & Dohme, Pfizer, and UCB and personal fees from AbbVie, Merck Sharp & Dohme, Hospira/Pfizer, Roche, UCB, Lilly, Hikma, Orion, Sanofi, Celltrion, Sandoz, Biogen, Amgen, Egis, Ewopharma, Mylan, EVA Pharma, and Gilead. Dr Bolstad reported receipt of personal fees from Roche, Janssen, and Novartis. Dr Haavardsholm reported receipt of personal fees from Pfizer, AbbVie, Celgene, Novartis, Janssen, Gilead, Lilly, and UCB. No other disclosures were reported.

Funding/Support: The study was funded by grants from the Norwegian Regional Health Authorities (interregional KLINBEFORSK grants) and the South-Eastern Norway Regional Health Authorities. The sponsor of the study was Diakonhjemmet Hospital.

Role of the Funder/Sponsor: Neither the funder of the study (the Regional Health Authorities) nor the sponsor (Diakonhjemmet Hospital) had any role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; or decision to submit the manuscript for publication. Neither the funder nor the sponsor had any right to veto publication or to control the decision regarding to which journal the manuscript was submitted. All drafts of the manuscript were prepared by the authors. All authors approved the final submitted version.

Data Sharing Statement: See Supplement 3.

Additional Contributions: We acknowledge the patient representatives, Jon Hagfors (Norwegian Rheumatism Association), Bjørn Gulbrandsen (Norwegian IBD Patient Organization), and Hilde Mellum (Psoriasis and Eczema Association of Norway), for their contributions during planning and conduct of the study. We acknowledge the members of the scientific advisory board, Josef Smolen, MD, PhD (University of Vienna), Alejandro C. Balsa, MD, PhD (University Hospital La Paz, Madrid), Geert D’Haens, MD, PhD (Amsterdam Academic Medical Center), Jørn Brynskov, MD, PhD (University of Copenhagen Herlev Hospital), Knut E. A. Lundin, MD, PhD (University of Oslo), and Diamant Thaci, MD, PhD (University of Lubeck). No compensation was received for their contributions to the protocol. We acknowledge Cecilie Moe, Bjørn Solvang, Nina Flatner, Trond Smedsrud, and Marius Eid, Department of Research Support for Clinical Trials at Oslo University Hospital, and Anja Bye, Clinical Research Unit Central Norway, for research support and data management (received compensation for their work).

References
1.
Maini  R , St Clair  EW , Breedveld  F ,  et al; ATTRACT Study Group.  Infliximab (chimeric anti-tumour necrosis factor alpha monoclonal antibody) versus placebo in rheumatoid arthritis patients receiving concomitant methotrexate: a randomised phase III trial.   Lancet. 1999;354(9194):1932-1939. doi:10.1016/S0140-6736(99)05246-0 PubMedGoogle ScholarCrossref
2.
Rutgeerts  P , Sandborn  WJ , Feagan  BG ,  et al.  Infliximab for induction and maintenance therapy for ulcerative colitis.   N Engl J Med. 2005;353(23):2462-2476. doi:10.1056/NEJMoa050516PubMedGoogle ScholarCrossref
3.
Hanauer  SB , Feagan  BG , Lichtenstein  GR ,  et al; ACCENT I Study Group.  Maintenance infliximab for Crohn’s disease: the ACCENT I randomised trial.   Lancet. 2002;359(9317):1541-1549. doi:10.1016/S0140-6736(02)08512-4PubMedGoogle ScholarCrossref
4.
Chaudhari  U , Romano  P , Mulcahy  LD , Dooley  LT , Baker  DG , Gottlieb  AB .  Efficacy and safety of infliximab monotherapy for plaque-type psoriasis: a randomised trial.   Lancet. 2001;357(9271):1842-1847. doi:10.1016/S0140-6736(00)04954-0 PubMedGoogle ScholarCrossref
5.
Thomas  SS , Borazan  N , Barroso  N ,  et al.  Comparative immunogenicity of TNF inhibitors: impact on clinical efficacy and tolerability in the management of autoimmune diseases. a systematic review and meta-analysis.   BioDrugs. 2015;29(4):241-258. doi:10.1007/s40259-015-0134-5PubMedGoogle ScholarCrossref
6.
Nencini  F , Vultaggio  A , Pratesi  S ,  et al.  The kinetics of antidrug antibodies, drug levels, and clinical outcomes in infliximab-exposed patients with immune-mediated disorders.   J Allergy Clin Immunol Pract. 2018;6(6):2065-2072. doi:10.1016/j.jaip.2018.04.007PubMedGoogle ScholarCrossref
7.
St Clair  EW , Wagner  CL , Fasanmade  AA ,  et al.  The relationship of serum infliximab concentrations to clinical improvement in rheumatoid arthritis: results from ATTRACT, a multicenter, randomized, double-blind, placebo-controlled trial.   Arthritis Rheum. 2002;46(6):1451-1459. doi:10.1002/art.10302PubMedGoogle ScholarCrossref
8.
Adedokun  OJ , Sandborn  WJ , Feagan  BG ,  et al.  Association between serum concentration of infliximab and efficacy in adult patients with ulcerative colitis.   Gastroenterology. 2014;147(6):1296-1307. doi:10.1053/j.gastro.2014.08.035PubMedGoogle ScholarCrossref
9.
Vande Casteele  N , Khanna  R , Levesque  BG ,  et al.  The relationship between infliximab concentrations, antibodies to infliximab and disease activity in Crohn’s disease.   Gut. 2 Gut. 2015;64(10):1539-1545. doi:10.1136/gutjnl-2014-307883PubMedGoogle ScholarCrossref
10.
Papamichael  K , Vogelzang  EH , Lambert  J , Wolbink  G , Cheifetz  AS .  Therapeutic drug monitoring with biologic agents in immune mediated inflammatory diseases.   Expert Rev Clin Immunol. 2019;15(8):837-848. doi:10.1080/1744666X.2019.1630273PubMedGoogle ScholarCrossref
11.
Medina  F , Plasencia  C , Goupille  P , Ternant  D , Balsa  A , Mulleman  D .  Current practice for therapeutic drug monitoring of biopharmaceuticals in rheumatoid arthritis.   Ther Drug Monit. 2017;39(4):364-369. doi:10.1097/FTD.0000000000000421PubMedGoogle ScholarCrossref
12.
Ma  C , Battat  R , Jairath  V , Vande Casteele  N .  Advances in therapeutic drug monitoring for small-molecule and biologic therapies in inflammatory bowel disease.   Curr Treat Options Gastroenterol. 2019;17(1):127-145. doi:10.1007/s11938-019-00222-9PubMedGoogle ScholarCrossref
13.
Grossberg  LB , Papamichael  K , Feuerstein  JD , Siegel  CA , Ullman  TA , Cheifetz  AS .  A survey study of gastroenterologists’ attitudes and barriers toward therapeutic drug monitoring of anti-TNF therapy in inflammatory bowel disease.   Inflamm Bowel Dis. 2017;24(1):191-197. doi:10.1093/ibd/izx023PubMedGoogle ScholarCrossref
14.
Smolen  JS , Landewé  R , Bijlsma  J ,  et al.  EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update.   Ann Rheum Dis. 2017;76(6):960-977. doi:10.1136/annrheumdis-2016-210715PubMedGoogle ScholarCrossref
15.
Vande Casteele  N , Herfarth  H , Katz  J , Falck-Ytter  Y , Singh  S .  American Gastroenterological Association Institute technical review on the role of therapeutic drug monitoring in the management of inflammatory bowel diseases.   Gastroenterology. 2017;153(3):835-857. doi:10.1053/j.gastro.2017.07.031PubMedGoogle ScholarCrossref
16.
Feuerstein  JD , Nguyen  GC , Kupfer  SS , Falck-Ytter  Y , Singh  S ; American Gastroenterological Association Institute Clinical Guidelines Committee.  American Gastroenterological Association Institute guideline on therapeutic drug monitoring in inflammatory bowel disease.   Gastroenterology. 2017;153(3):827-834. doi:10.1053/j.gastro.2017.07.032PubMedGoogle ScholarCrossref
17.
Ricciuto  A , Dhaliwal  J , Walters  TD , Griffiths  AM , Church  PC .  Clinical outcomes with therapeutic drug monitoring in inflammatory bowel disease: a systematic review with meta-analysis.   J Crohns Colitis. 2018;12(11):1302-1315. doi:10.1093/ecco-jcc/jjy109PubMedGoogle ScholarCrossref
18.
World Medical Association.  World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects.   JAMA. 2013;310(20):2191-2194. doi:10.1001/jama.2013.281053Google ScholarCrossref
19.
Syversen  SW , Goll  GL , Jørgensen  KK ,  et al.  Therapeutic drug monitoring of infliximab compared to standard clinical treatment with infliximab: study protocol for a randomised, controlled, open, parallel-group, phase IV study (the NOR-DRUM study).   Trials. 2020;21(1):13. doi:10.1186/s13063-019-3734-4PubMedGoogle ScholarCrossref
20.
Prevoo  ML , van ’t Hof  MA , Kuper  HH , van Leeuwen  MA , van de Putte  LB , van Riel  PL .  Modified disease activity scores that include twenty-eight-joint counts: development and validation in a prospective longitudinal study of patients with rheumatoid arthritis.   Arthritis Rheum. 1995;38(1):44-48. doi:10.1002/art.1780380107PubMedGoogle ScholarCrossref
21.
England  BR , Tiong  BK , Bergman  MJ ,  et al.  2019 update of the American College of Rheumatology recommended rheumatoid arthritis disease activity measures.   Arthritis Care Res (Hoboken). 2019;71(12):1540-1555. doi:10.1002/acr.24042PubMedGoogle ScholarCrossref
22.
Lukas  C , Landewé  R , Sieper  J ,  et al; Assessment of Spondyloarthritis International Society.  Development of an ASAS-endorsed disease activity score (ASDAS) in patients with ankylosing spondylitis.   Ann Rheum Dis. 2009;68(1):18-24. doi:10.1136/ard.2008.094870PubMedGoogle ScholarCrossref
23.
Machado  P , Landewé  R , Lie  E ,  et al; Assessment of Spondyloarthritis International Society.  Ankylosing Spondylitis Disease Activity Score (ASDAS): defining cut-off values for disease activity states and improvement scores.   Ann Rheum Dis. 2011;70(1):47-53. doi:10.1136/ard.2010.138594PubMedGoogle ScholarCrossref
24.
Lewis  JD , Chuai  S , Nessel  L , Lichtenstein  GR , Aberra  FN , Ellenberg  JH .  Use of the noninvasive components of the Mayo score to assess clinical response in ulcerative colitis.   Inflamm Bowel Dis. 2008;14(12):1660-1666. doi:10.1002/ibd.20520PubMedGoogle ScholarCrossref
25.
Harvey  RF , Bradshaw  JM .  A simple index of Crohn’s-disease activity.   Lancet. 1980;1(8167):514. doi:10.1016/S0140-6736(80)92767-1PubMedGoogle ScholarCrossref
26.
Vermeire  S , Schreiber  S , Sandborn  WJ , Dubois  C , Rutgeerts  P .  Correlation between the Crohn’s disease activity and Harvey-Bradshaw indices in assessing Crohn’s disease severity.   Clin Gastroenterol Hepatol. 2010;8(4):357-363. doi:10.1016/j.cgh.2010.01.001PubMedGoogle ScholarCrossref
27.
Fredriksson  T , Pettersson  U .  Severe psoriasis—oral therapy with a new retinoid.   Dermatologica. 1978;157(4):238-244. doi:10.1159/000250839PubMedGoogle ScholarCrossref
28.
Jørgensen  KK , Olsen  IC , Goll  GL ,  et al; NOR-SWITCH Study Group.  Switching from originator infliximab to biosimilar CT-P13 compared with maintained treatment with originator infliximab (NOR-SWITCH): a 52-week, randomised, double-blind, non-inferiority trial.   Lancet. 2017;389(10086):2304-2316. doi:10.1016/S0140-6736(17)30068-5PubMedGoogle ScholarCrossref
29.
D’Haens  G , Sandborn  WJ , Feagan  BG ,  et al.  A review of activity indices and efficacy end points for clinical trials of medical therapy in adults with ulcerative colitis.   Gastroenterology. 2007;132(2):763-786. doi:10.1053/j.gastro.2006.12.038PubMedGoogle ScholarCrossref
30.
Berth-Jones  J , Grotzinger  K , Rainville  C ,  et al.  A study examining inter- and intrarater reliability of three scales for measuring severity of psoriasis: Psoriasis Area and Severity Index, Physician’s Global Assessment and Lattice System Physician’s Global Assessment.   Br J Dermatol. 2006;155(4):707-713. doi:10.1111/j.1365-2133.2006.07389.xPubMedGoogle ScholarCrossref
31.
Yoo  DH , Hrycaj  P , Miranda  P ,  et al.  A randomised, double-blind, parallel-group study to demonstrate equivalence in efficacy and safety of CT-P13 compared with innovator infliximab when coadministered with methotrexate in patients with active rheumatoid arthritis: the PLANETRA study.   Ann Rheum Dis. 2013;72(10):1613-1620. doi:10.1136/annrheumdis-2012-203090PubMedGoogle ScholarCrossref
32.
D’Haens  G , Vermeire  S , Lambrecht  G ,  et al; GETAID.  Increasing infliximab dose based on symptoms, biomarkers, and serum drug concentrations does not increase clinical, endoscopic, and corticosteroid-free remission in patients with active luminal Crohn’s disease.   Gastroenterology. 2018;154(5):1343-1351. doi:10.1053/j.gastro.2018.01.004PubMedGoogle ScholarCrossref
33.
Vande Casteele  N , Ferrante  M , Van Assche  G ,  et al.  Trough concentrations of infliximab guide dosing for patients with inflammatory bowel disease.   Gastroenterology. 2015;148(7):1320-1329. doi:10.1053/j.gastro.2015.02.031PubMedGoogle ScholarCrossref
34.
Teresa  J , Chamaida  PR , Ana  MF ,  et al.  Predictive value of serum infliximab levels at induction phase in rheumatoid arthritis patients.   Open Rheumatol J. 2017;11:75-87. doi:10.2174/1874312901711010075PubMedGoogle ScholarCrossref
35.
Kobayashi  T , Suzuki  Y , Motoya  S ,  et al.  First trough level of infliximab at week 2 predicts future outcomes of induction therapy in ulcerative colitis—results from a multicenter prospective randomized controlled trial and its post hoc analysis.   J Gastroenterol. 2016;51(3):241-251. doi:10.1007/s00535-015-1102-zPubMedGoogle ScholarCrossref
36.
Cornillie  F , Hanauer  SB , Diamond  RH ,  et al.  Postinduction serum infliximab trough level and decrease of C-reactive protein level are associated with durable sustained response to infliximab: a retrospective analysis of the ACCENT I trial.   Gut. 2014;63(11):1721-1727. doi:10.1136/gutjnl-2012-304094PubMedGoogle ScholarCrossref
37.
Papamichael  K , Van Stappen  T , Vande Casteele  N ,  et al.  Infliximab concentration thresholds during induction therapy are associated with short-term mucosal healing in patients with ulcerative colitis.   Clin Gastroenterol Hepatol. 2016;14(4):543-549. doi:10.1016/j.cgh.2015.11.014PubMedGoogle ScholarCrossref
38.
Verstockt  B , Moors  G , Bian  S ,  et al.  Influence of early adalimumab serum levels on immunogenicity and long-term outcome of anti-TNF naive Crohn’s disease patients: the usefulness of rapid testing.   Aliment Pharmacol Ther. 2018;48(7):731-739. doi:10.1111/apt.14943PubMedGoogle ScholarCrossref
39.
Kennedy  NA , Heap  GA , Green  HD ,  et al; UK Inflammatory Bowel Disease Pharmacogenetics Study Group.  Predictors of anti-TNF treatment failure in anti-TNF-naive patients with active luminal Crohn’s disease: a prospective, multicentre, cohort study.   Lancet Gastroenterol Hepatol. 2019;4(5):341-353. doi:10.1016/S2468-1253(19)30012-3PubMedGoogle ScholarCrossref
40.
Mrowietz  U , Kragballe  K , Reich  K ,  et al.  Definition of treatment goals for moderate to severe psoriasis: a European consensus.   Arch Dermatol Res. 2011;303(1):1-10. doi:10.1007/s00403-010-1080-1PubMedGoogle ScholarCrossref
41.
Steenholdt  C , Bendtzen  K , Brynskov  J , Thomsen  OØ , Ainsworth  MA .  Clinical implications of measuring drug and anti-drug antibodies by different assays when optimizing infliximab treatment failure in Crohn’s disease: post hoc analysis of a randomized controlled trial.   Am J Gastroenterol. 2014;109(7):1055-1064. doi:10.1038/ajg.2014.106PubMedGoogle ScholarCrossref
AMA CME Accreditation Information

Credit Designation Statement: The American Medical Association designates this Journal-based CME activity activity for a maximum of 1.00  AMA PRA Category 1 Credit(s)™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

Successful completion of this CME activity, which includes participation in the evaluation component, enables the participant to earn up to:

  • 1.00 Medical Knowledge MOC points in the American Board of Internal Medicine's (ABIM) Maintenance of Certification (MOC) program;;
  • 1.00 Self-Assessment points in the American Board of Otolaryngology – Head and Neck Surgery’s (ABOHNS) Continuing Certification program;
  • 1.00 MOC points in the American Board of Pediatrics’ (ABP) Maintenance of Certification (MOC) program;
  • 1.00 Lifelong Learning points in the American Board of Pathology’s (ABPath) Continuing Certification program; and
  • 1.00 CME points in the American Board of Surgery’s (ABS) Continuing Certification program

It is the CME activity provider's responsibility to submit participant completion information to ACCME for the purpose of granting MOC credit.

Close
Want full access to the AMA Ed Hub?
After you sign up for AMA Membership, make sure you sign in or create a Physician account with the AMA in order to access all learning activities on the AMA Ed Hub
Buy this activity
Close
Want full access to the AMA Ed Hub?
After you sign up for AMA Membership, make sure you sign in or create a Physician account with the AMA in order to access all learning activities on the AMA Ed Hub
Buy this activity
Close
With a personal account, you can:
  • Access free activities and track your credits
  • Personalize content alerts
  • Customize your interests
  • Fully personalize your learning experience
Education Center Collection Sign In Modal Right
Close

Name Your Search

Save Search
With a personal account, you can:
  • Access free activities and track your credits
  • Personalize content alerts
  • Customize your interests
  • Fully personalize your learning experience
Close
Close

Lookup An Activity

or

My Saved Searches

You currently have no searches saved.

Close

My Saved Courses

You currently have no courses saved.

Close