Abstract
Evidence-based medicine is widely promoted for decision-making in health care and is associated with improved patient outcomes. Critics have suggested that evidence-based medicine focuses primarily on groups of patients rather than individuals, but often fail to consider subgroup analyses, N-of-1 trials, and the incorporation of patient values and preferences. Precision medicine has been promoted as an approach to individualize diagnosis and treatment of diseases through genetic, biomarker, phenotypic, and psychosocial characteristics. However, there are often high costs associated with personalized medicine, and high-quality evidence is lacking for effectiveness in many applications. For the potential of personalized medicine to be realized, it must adhere to the principles of evidence-based medicine: (1) evidence in isolation is not sufficient to make clinical decisions—patient’s values and preferences as well as resource implications must be considered, and (2) there is a hierarchy of evidence to guide clinical decision-making and studies at lower risk of bias are likely to provide more trustworthy findings.
Keywords: evidence-based medicine, precision medicine, decision-making
Evidence-based medicine
Evidence-based medicine (EBM) represents a paradigm for clinical practice that evolved out of a need for greater objectivity in clinical decision-making. EBM is defined as the “conscientious, explicit, and judicious use of current best evidence in making decisions about the care of individual patients,” rather than making clinical decisions solely on clinical experience and pathophysiologic rationale.1,2 EBM is now widely accepted as optimal practice for decision-making in health care.3
EBM is based on three fundamental principles. First, there is a hierarchy of evidence based on study design—from approaches that are at lower risk of bias (e.g. rigorously conducted randomized controlled trials) to approaches that are at higher risk of bias (e.g. observational studies). Second, informed clinical decision-making requires use of all best available evidence, usually from systematic reviews to avoid selection bias. A notable resource is the Cochrane Collaboration, which provides reviews of evidence from comparative research.4 Third, evidence alone is never enough for clinical decision-making, and clinicians must also consider patient’s values and preferences.
The application of EBM has been shown to result in better outcomes for patients. For example, the development of the British Thoracic Society’s 1990 asthma guidelines led to increased prescription of inhaled steroids and use of personal care plans, and subsequently led to decreased morbidity and mortality rates.5–7 Another example is the UK National Institute for Health and Care Excellence guidelines for prevention of venous thromboembolism following surgery, which led to reductions in thromboembolic complications.8
Purported limitations of EBM
Average effects vs. the proportion that benefit
While EBM provides many important benefits to clinical decision-making, it is not without limitations. Some have criticized EBM for focusing on groups of patients rather than on the individual.9,10 Specifically, when trialists report evidence for treatment efficacy, the results are often based on the average treatment effect and do not apply to all patients. However, guidance exists for reporting the proportion of patients that experience important benefit, instead of focusing only on average effects.11 For example, high-quality evidence from 27 studies (13 876 patients) supports the notion that opioids versus placebo provide a small improvement in pain for patients with chronic non-cancer pain—an average reduction of 0.64 cm on a 10 cm visual analogue scale for pain.12 This effect is smaller than the minimally important difference (MID), the smallest change in an instrument score that patients perceive is important, of 1 cm.13 If every patient experienced this same effect, opioids should not be used for analgesia in this population; however, the application of methods to calculate the proportion of patients that achieve the MID results in a risk difference of 11%, meaning that 11% more patients with chronic non-cancer pain treated with opioids will achieve important pain relief versus those who received placebo. This translates to a number need to treat (NNT) of nine, meaning that nine patients need to receive treatment to achieve an important benefit in one patient.
Subgroup effects
When pooling results across trials in a meta-analysis, there may be heterogeneity in the treatment effect, which suggests that there may be subgroups of patients (i.e. older, sicker) that have a different response to treatment or vulnerability to adverse effects.14 EBM has recognized this issue, and provides strategies for exploring possible subgroup effects to guide treatment of individuals with important prognostic factors;15,16 however, many reported subgroup effects fail to meet criteria for validity.17
When credible, clinicians can determine the baseline risk relevant to subgroups of patients, and calculate the expected effect of an intervention by multiplying their baseline risk by the relative risk.17 For example, consider a patient with a disease that, on average, is associated with a 1% risk of death over the next year, and administration of a certain drug versus placebo has shown a 20% relative risk reduction of death. This equates to an absolute risk reduction (ARR) of 0.2% (1% × 0.2 = 0.2%), or a NNT of 500 (100/0.2 = 500), meaning 500 patients need to be treated with the drug to prevent one death. Now consider a subgroup of patients (e.g. those with more severe disease burden) who have a risk of death over the next year of 5%—their ARR with treatment would be 1% (5% × 0.2 = 1%). This translates to a NNT of 100 (100/1.0 = 100), meaning 100 patients need to be treated with the drug to prevent one death.
N-of-1 trials
Randomized controlled trials (RCTs) are the most methodologically rigorous study design to establish evidence of treatment efficacy; however, the results are only generalizable to patients that resemble the study population. To maintain methodological safeguards against risk of bias in RCTs (such as random sequence generation, allocation concealment, and blinding) and to ensure applicability to individual patients, N-of-1 RCTs have been proposed for evaluating treatment effects in individuals.18,19 In such trials, the experimental intervention and control (or a competing therapy) are administered in pairs and ordered randomly to confirm the effectiveness of treatment among individual patients.18 Treatments are separated by a washout period, a designated period of time when a participant is taken off a study intervention to eliminate the effects of the treatment. The number of pairs of interventions typically varies from two to seven, but the clinician and patient can decide to stop when they establish that there are, or are not, important differences between interventions (Fig. 1).
Figure 1.
Basic design for N-of-1 randomized controlled trial.
Precision medicine
Precision medicine (PM), otherwise known as personalized or individualized medicine, tailors the diagnosis and treatment of diseases to the individual based on genetic, biomarker, phenotypic, or psychosocial characteristics; in other words, it is the concept of administering the right treatment, to the right patient, at the right time.20 The recent completion of the Human Genome Project, along with technological advances for characterizing patients using proteomics, metabolomics, and genomics, provides a unique and exciting opportunity for PM to play an important role in clinical decision-making.21 Proponents of PM suggest it has the potential to re-focus medicine from reaction to prevention, direct the selection of optimal therapy, improve quality of life, reduce adverse drug reactions, increase treatment adherence, and reduce overall health care expenses.22,23
Shift from reaction to prevention
The field of oncology holds great promise for the application of PM as a result of increased understanding of oncogenic mechanisms.21 For example, women with certain BRCA1 and BRCA2 gene mutations have a 72% and 69% risk of developing breast cancer, and a 44% and 17% risk of developing ovarian cancer, respectively.24 Furthermore, the molecular diagnosis of germ-lines re-arranged during transfection mutations in individuals with multiple endocrine neoplasia type 2 allows for codon-directed prophylactic thyroidectomy and regular screening for pheochromocytoma, medullary thyroid cancer, and hyperparathyroidism.25 These advancements in technology enable clinicians to identify at-risk individuals with genetic tests, and promote preventive measures, such as increased frequency of imaging, chemoprevention, and prophylactic surgery.22
Direct the selection of optimal therapy
Up to 50% of patients do not respond to initial treatment for diseases such as arthritis, diabetes, asthma, or depression.26 It has been suggested that, in some cases, differences in response to treatment are related to mutations in genes that code for drug-metabolizing enzymes, drug targets, or drug transporters.27–29 For example, diagnostic tests are commonly used to determine which breast tumors over-express the human epidermal growth factor receptor type 2 (HER2), a biomarker that is associated with worse prognosis but also predicts a better response to trastuzumab—a monoclonal antibody.30 Moreover, an estimated 40% of patients with metastatic colon cancer do not respond to cetuximab and panitumumab because of mutations of the KRAS gene.31 This discovery led to recommendations that only patients without mutations of the KRAS gene should be treated with cetuximab and panitumumab.32
Reduce adverse drug reactions and increase adherence to treatment
It has been estimated that up to 5.3% of all hospital admissions are related to adverse drug reactions.33 Many adverse drug reactions result from variations in genes that code for drug-metabolizing enzymes, such as cytochrome P450 (CYP450), which can result in drugs being metabolized either slower or faster than normal.34,35 As a result, some patients may have difficulty eliminating certain drugs, leading to potential overdose toxicity, while others may eliminate drugs before they are able to have an effect. For example, 5-8% of HIV patients managed with abacavir may experience multi-organ system hypersensitivity because of presence of the HLA-B*5701 gene.22 This adverse reaction can be fatal in some cases, which has now prompted genetic testing for almost all HIV patients receiving abacavir. Reducing potential adverse drug reactions through genetic testing is one way to improve patient adherence to treatment. Another way to improve adherence is through knowledge of genetic predisposition to a condition. For example, patients who screen positive for predisposition to familial hypercholesterolemia, and are made aware of this, have a treatment adherence to lipid-lowering medication of 86% after 2 years, compared with 38% prior to testing.36
Limitations of PM
Limited evidence of clinical benefit
Although the promise of PM is enticing, and broad implementation of multiplex hotspot testing is feasible, only 13-40% of patients enrolled into genotype-matched trials have presented with actionable alterations, which risks attenuation of treatment effects.37–40 With this in mind, the current evidence suggests that clinical benefits of biomarker-based treatment strategies may be limited.41,42 For example, a 2016 systematic review of 346 studies that compared phase 1 cancer drug trials with biomarker-based treatment strategies to trials without this approach concluded that a personalized approach resulted in a median progression-free survival of 5.7 months (95% CI 2.6–13.8) versus 2.95 months (95% CI 2.3–3.7).41 This review, however, did not assess risk of bias of individuals trials, or the overall quality of evidence for the outcomes they reported on, and was unable to assess effects on overall survival because of insufficient data.
Limitations of biomarkers and molecular targeted drugs
The diagnostic accuracy of genetic tests is limited, and not all genetic markers have clinical significance. For example, there are reported cases in which women have undergone unnecessary removal of their ovaries after receiving false positive results of genetic testing.20 There is a great need for better biomarkers to assist with the diagnosis of diseases to help guide optimal treatment. Furthermore, even if accurate genetic tests are established, molecular targeted drugs must be developed that are able to successfully target signaling pathways. Available molecular targeted drugs only partially inhibit signaling pathways and may be too toxic to be used in combination. In addition, although some drugs can target signaling pathways in cancer patients, cancer cells have the capacity to develop a resistance to them by up-regulating the pathway or activation of alternative pathways.43,44
Although the above examples largely focus on genetic information to guide PM, this approach also makes use of differences in patient’s biomarkers, environment, and lifestyle to customize care. Preventive or therapeutic interventions can then be offered to those who are most likely to benefit, sparing expense and side effects for those who will not.
Policy challenges and costs
There are policy challenges to the widespread uptake of PM, such as the regulation of genetic tests in such a way that encourages innovation but also protects patient confidentiality.20,45 Health and drug regulatory authorities need to establish clear guidelines for the identification and approval of personalized drugs and their related diagnostic tests for clinical use.20,22 Furthermore, the costs of developing and marketing new molecular targeted drugs are high, and may divert resources from the development of more clinically effective drugs. If health and regulatory authorities are to fund PM research, there should be independent assessors who regularly appraise the cost-benefit ratio of targeted drugs.46 Until there are more studies demonstrating clinical effectiveness of molecular targeted drugs, it may be difficult to justify their high costs.
Conclusions
While EBM and PM have their own merits and limitations, these approaches complement rather than oppose one another. The promise of personalized patient care is powerful and has the potential to fundamentally change health care; however, more high-quality evidence is needed to guide the application of PM to areas in which the benefits outweigh the harms.
References
- 1. Sackett DL, Rosenberg WM, Gray JA, et al. Evidence based medicine: what it is and what it isn’t. BMJ 1996;312:71–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Evidence Based Medicine Working Group . Evidence-based medicine: a new approach to teaching the practice of medicine. JAMA 1992;268:2420–5. [DOI] [PubMed] [Google Scholar]
- 3. Fernandez A, Sturmberg J, Lukersmith S, et al. Evidence-based medicine: is it a bridge too far? Health Res Policy Syst 2015;13:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Sheridan DJ, Julian DG. Achievements and Limitations of Evidence-Based Medicine. J Am Coll Cardiol 2016;68:204–13. [DOI] [PubMed] [Google Scholar]
- 5. British Thoracic Society . Guidelines for management of asthma in adults: I. Chronic persistent asthma. BMJ 1990;301:651–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Majeed A, Ferguson J, Field J. Prescribing of beta-2 agonists and inhaled steroids in England: trends between 1992 and 1998, and association with material deprivation, chronic illness and asthma mortality rates. J Public Health Med 1999;21:395–400. [DOI] [PubMed] [Google Scholar]
- 7. Kelly MP, Capewell S. Relative contributions of changes in risk factors and treatment to the reduction in coronary heart disease mortality. London, UK: Health Development Agency, 2004. [Google Scholar]
- 8. Lau BD, Haut ER. Practices to prevent venous thromboembolism: a brief review. BMJ Qual Saf 2014;23:187–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Bensing J. Bridging the gap. The separate worlds of evidence-based medicine and patient-centered medicine. Patient Educ Couns 2000;39:17–25. [DOI] [PubMed] [Google Scholar]
- 10. Groopman J. How doctors think. Boston, Massachusetts, USA: Houghton Mifflin Company, 2007. [Google Scholar]
- 11. Busse JW, Bartlett S, Dougados M, et al. Optimal Strategies for Reporting Pain in Clinical Trials and Systematic Reviews: Recommendations from a 2014 OMERACT Workshop. J Rheumatol 2015;42:1962–70. [DOI] [PubMed] [Google Scholar]
- 12. Busse JW, Craigie S, Juurlink DN, et al. The 2017 Canadian Guideline for Opioid Therapy and Chronic Non-Cancer Pain. CMAJ 2017;189:E659–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Kelly AM. The minimum clinically significant difference in visual analogue scale pain score does not differ with severity of pain. Emerg Med J 2001;18:205–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Kravitz RL, Duan N, Braslow J. Evidence-based medicine, heterogeneity of treatment effects, and the trouble with averages. Milbank Q 2004;82:661–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Sun X, Briel M, Walter SD, et al. Is a subgroup effect believable? Updating criteria to evaluate the credibility of subgroup analyses. BMJ 2010;340:c117. [DOI] [PubMed] [Google Scholar]
- 16. Straus SE. Individualizing treatment decisions. The likelihood of being helped or harmed. Eval Health Prof 2002;25:210–24. [DOI] [PubMed] [Google Scholar]
- 17. Sun X, Briel M, Busse JW, et al. Credibility of claims of subgroup effects in randomized controlled trials: systematic review. BMJ 2012;344:e1553. [DOI] [PubMed] [Google Scholar]
- 18. Guyatt GH, Sackett D, Taylor DW, et al. Determining optimal therapy—randomized trials in individual patients. N Engl J Med 1986;314:889–92. [DOI] [PubMed] [Google Scholar]
- 19. Guyatt GH, Keller JL, Jaeschke R, et al. The n-of-1 randomized controlled trial: clinical usefulness. Our three-year experience. Ann Intern Med 1990;112:293–9. [DOI] [PubMed] [Google Scholar]
- 20. Hamburg MA, Collins FS. The path to personalized medicine. N Engl J Med 2010;363:301–4. [DOI] [PubMed] [Google Scholar]
- 21. Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med 2015;372:793–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Personalized Medicine Coalition . The case for personalized medicine. Personalized Medicine Coalition: 2014. (Available at: http://www.personalizedmedicinecoalition.org/Resources/The_Case_for_Personalized_Medicine)
- 23. Milani L, Leitsalu L, Metspalu A. An epidemiological perspective of personalized medicine: the Estonian experience. J Intern Med 2015;277:188–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Kuchenbaecker KB, Hopper JL, Barnes DR, et al. Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA 2017;317:2402–16. [DOI] [PubMed] [Google Scholar]
- 25. Moore FD, Dluhy RG. Prophylactic thyroidectomy in MEN-2A—a stitch in time? N Engl J Med 2005;353:1162–4. [DOI] [PubMed] [Google Scholar]
- 26. Spear BB, Heath-Chiozzi M, Huff J. Clinical application of pharmacogenetics. Trends Mol Med 2001;7:201–4. [DOI] [PubMed] [Google Scholar]
- 27. Mangravite LM, Thorn CF, Krauss RM. Clinical implications of pharmacogenomics of statin treatment. Pharmacogenomics J 2006;6:360–74. [DOI] [PubMed] [Google Scholar]
- 28. Rieder MJ, Reiner AP, Gage BF, et al. Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. N Engl J Med 2005;352:2285–93. [DOI] [PubMed] [Google Scholar]
- 29. Terra SG, Hamilton KK, Pauly DF, et al. Beta1-adrenergic receptor polymorphisms and left ventricular remodeling changes in response to beta-blocker therapy. Pharmacogenet Genomics 2005;15:227–34. [DOI] [PubMed] [Google Scholar]
- 30. Onitilo AA, Engel JM, Greenlee RT, et al. Breast Cancer Subtypes Based on ER/PR and Her2 Expression: Comparison of Clinicopathologic Features and Survival. Clin Med Res 2009;7:4–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Lièvre A, Bachet JB, Le Corre D, et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res 2006;66:3992–5. [DOI] [PubMed] [Google Scholar]
- 32. National Comprehensive Cancer Network . Guidelines in Oncology: Colon Cancer. v.2.2009. NCCN web site. Available at: http://www.nccn.org/professionals/physician_gls/PDF/colon.pdf [DOI] [PubMed]
- 33. Kongkaew C, Noyce PR, Ashcroft DM. Hospital admissions associated with adverse drug reactions: a systematic review of prospective observational studies. Ann Pharmacother 2008;42:1017–25. [DOI] [PubMed] [Google Scholar]
- 34. Phillips KA, Veenstra DL, Oren E, et al. Potential role of pharmacogenomics in reducing adverse drug reactions: a systematic review. JAMA 2001;286:2270–9. [DOI] [PubMed] [Google Scholar]
- 35. Blue Cross Blue Shield Technology Evaluation Center . Special Report: Genotyping for Cytochrome P450 Polymorphisms To Determine Drug-Metabolizer Status. Assess Program 2004;19:1–34. [PubMed] [Google Scholar]
- 36. Umans-Eckenhausen MA, Defesche JC, van Dam MJ, et al. Long-term compliance with lipid-lowering medication after genetic screening for familial hypercholesterolemia. Arch Intern Med 2003;163:658. [DOI] [PubMed] [Google Scholar]
- 37. André F, Bachelot T, Commo F, et al. Comparative genomic hybridisation array and DNA sequencing to direct treatment of metastatic breast cancer: a multicentre, prospective trial (SAFIR01/UNICANCER). Lancet Oncol 2014;15:267–74. [DOI] [PubMed] [Google Scholar]
- 38. Kris MG, Johnson BE, Berry LD, et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA 2014;311:1998–2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Meric-Bernstam F, Brusco L, Shaw K, et al. Feasibility of large-scale genomic testing to facilitate enrollment onto genomically matched clinical trials. J Clin Oncol 2015;33:2753–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Stockley TL, Oza AM, Berman HK, et al. Molecular profiling of advanced solid tumors and patient outcomes with genotype-matchedclinical trials: the Princess Margaret IMPACT/COMPACT trial. Genome Med 2016;8:109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Schwaederle M, Zhao M, Lee JJ, et al. Association of Biomarker-Based Treatment Strategies With Response Rates and Progression-Free Survival in Refractory Malignant Neoplasms: A Meta-analysis. JAMA Oncol 2016;2:1452–9. [DOI] [PubMed] [Google Scholar]
- 42. Prasad V. The precision oncology illusion. Nature 2016;537:S63. [DOI] [PubMed] [Google Scholar]
- 43. Johnson GL, Stuhlmiller TJ, Angus SP, et al. Molecular pathways: adaptive kinome reprogramming in response to targeted inhibition of the BRAF-MEK-ERK pathway in cancer. Clin Cancer Res 2014;20:2516–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Klein ME, Parvez MM, Shin JG. Clinical Implementation of Pharmacogenomics for Personalized Precision Medicine: Barriers and Solutions. J Pharm Sci 2017;106:2368–79. [DOI] [PubMed] [Google Scholar]
- 45. Tannock IF, Hickman JA. Limits to Personalized Cancer Medicine. N Engl J Med 2016;375:1289–94. [DOI] [PubMed] [Google Scholar]
- 46. Joyner MJ, Paneth N, Ioannidis JP. What Happens When Underperforming Big Ideas in Research Become Entrenched? JAMA 2016;316:1355–6. [DOI] [PubMed] [Google Scholar]