cambridge university press evidence based diagnosis feb 2009 kho tài liệu bách khoa

311 121 0
 cambridge university press evidence based diagnosis feb 2009 kho tài liệu bách khoa

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

This page intentionally left blank Evidence-Based Diagnosis Evidence-Based Diagnosis is a textbook about diagnostic, screening, and prognostic tests in clinical medicine The authors’ approach is based on many years of experience teaching physicians in a clinical research training program Although requiring only a minimum of mathematics knowledge, the quantitative discussions in this book are deeper and more rigorous than those in most introductory texts The book includes numerous worked examples and 60 problems (with answers) based on real clinical situations and journal articles The book will be helpful and accessible to anyone looking to select, develop, or market medical tests Topics covered include: r The diagnostic process r Test reliability and accuracy r Likelihood ratios r ROC curves r Testing and treatment thresholds r Critical appraisal of studies of diagnostic, screening, and prognostic tests r Test independence and methods of combining tests r Quantifying treatment benefits using randomized trials and observational studies r Bayesian interpretation of P-values and confidence intervals r Challenges for evidence-based diagnosis Thomas B Newman is Chief of the Division of Clinical Epidemiology and Professor of Epidemiology and Biostatistics and Pediatrics at the University of California, San Francisco He previously served as Associate Director of the UCSF/Stanford Robert Wood Johnson Clinical Scholars Program and Associate Professor in the Department of Laboratory Medicine at UCSF He is a co-author of Designing Clinical Research and a practicing pediatrician Michael A Kohn is Associate Clinical Professor of Epidemiology and Biostatistics at the University of California, San Francisco, where he teaches clinical epidemiology and evidence-based medicine He is also an emergency physician with more than 20 years of clinical experience, currently practicing at Mills–Peninsula Medical Center in Burlingame, California Evidence-Based Diagnosis Thomas B Newman University of California, San Francisco Michael A Kohn University of California, San Francisco CAMBRIDGE UNIVERSITY PRESS Cambridge, New York, Melbourne, Madrid, Cape Town, Singapore, São Paulo Cambridge University Press The Edinburgh Building, Cambridge CB2 8RU, UK Published in the United States of America by Cambridge University Press, New York www.cambridge.org Information on this title: www.cambridge.org/9780521886529 © Thomas B Newman and Michael A Kohn 2009 This publication is in copyright Subject to statutory exception and to the provision of relevant collective licensing agreements, no reproduction of any part may take place without the written permission of Cambridge University Press First published in print format 2009 ISBN-13 978-0-511-47937-3 eBook (EBL) ISBN-13 978-0-521-88652-9 hardback ISBN-13 978-0-521-71402-0 paperback Cambridge University Press has no responsibility for the persistence or accuracy of urls for external or third-party internet websites referred to in this publication, and does not guarantee that any content on such websites is, or will remain, accurate or appropriate Contents Preface Acknowledgments & Dedication Abbreviations/Acronyms Introduction: understanding diagnosis and diagnostic testing Reliability and measurement error 10 Dichotomous tests 39 Multilevel and continuous tests 68 Critical appraisal of studies of diagnostic tests 94 Screening tests 116 Prognostic tests and studies 138 Multiple tests and multivariable decision rules 156 Quantifying treatment effects using randomized trials 186 10 Alternatives to randomized trials for estimating treatment effects 206 11 Understanding P-values and confidence intervals 220 12 Challenges for evidence-based diagnosis 239 Answers to problems Index v page vii ix xi 255 287 Preface This is a book about diagnostic testing It is aimed primarily at clinicians, particularly those who are academically minded, but it should be helpful and accessible to anyone involved with selection, development, or marketing of diagnostic, screening, or prognostic tests Although we admit to a love of mathematics, we have restrained ourselves and kept the math to a minimum – a little simple algebra and only three Greek letters, κ (kappa), α (alpha), and β (beta) Nonetheless, quantitative discussions in this book go deeper and are more rigorous than those typically found in introductory clinical epidemiology or evidence-based medicine texts Our perspective is that of skeptical consumers of tests We want to make proper diagnoses and not miss treatable diseases Yet, we are aware that vast resources are spent on tests that too frequently provide wrong answers or right answers of little value, and that new tests are being developed, marketed, and sold all the time, sometimes with little or no demonstrable or projected benefit to patients This book is intended to provide readers with the tools they need to evaluate these tests, to decide if and when they are worth doing, and to interpret the results The pedagogical approach comes from years of teaching this material to physicians, mostly Fellows and junior faculty in a clinical research training program We have found that many doctors, including the two of us, can be impatient when it comes to classroom learning We like to be shown that the material is important and that it will help us take better care of our patients, understand the literature, and improve our research For this reason, in this book we emphasize real-life examples When we care for patients and read journal articles, we frequently identify issues that the material we teach can help people understand We have decided what material to include in this book largely by creating homework problems from patients and articles we have encountered, and then making sure that we covered in the text the material needed to solve them This explains the disproportionate number of pediatric and emergency medicine examples, and the relatively large portion of the book devoted to problems and answers – the parts we had the most fun writing vii viii Preface Although this is primarily a book about diagnosis, two of the twelve chapters are about evaluating treatments – both using randomized trials (Chapter 9) and observational studies (Chapter 10) The reason is that evidence-based diagnosis requires not only being able to evaluate tests and the information they provide, but also the value of that information – how it will affect treatment decisions, and how those decisions will affect patients’ health For this reason, the chapters about treatments emphasize quantifying risks and benefits Other reasons for including the material about treatments, which also apply to the material about P-values and confidence intervals in Chapter 11, are that we love to teach it, have lots of good examples, and are able to focus on material neglected (or even wrong) in other books After much deliberation, we decided to include in this text answers to all of the problems However, we strongly encourage readers to think about and even write out the answers to the problems before looking at the answers at the back of the book The disadvantage of including all of the answers is that instructors wishing to use this book for a course will have to create new problems for any take-home or open-book examinations Because that includes us, we will continue to write new problems, and will be happy to share them with others who are teaching courses based on this book We will post the additional problems on the book’s Web site: http://www epibiostat.ucsf.edu/ebd Several of the problems in this book are adapted from problems our students created in our annual final examination problem-writing contest Similarly, we encourage readers to create problems and share them with us With your permission, we will adapt them for the second edition! 281 Chapter Problem answers: quantifying treatment effects 3b Death or liver transplant No death or liver transplant Total 95 48 103 53 Interferon Alpha Untreated RC = 5/53 = 0.094 RT = 8/103 = 0.078 This difference in risk 5/53 vs 8/103 is totally consistent with chance (P = 0.72) RR = 0.078/0.094 = 0.82 (95% CI 0.28 - 2.39) 3c The conclusion is technically correct, but the abstract is potentially misleading Many readers will think that this study suggests that treatment with alpha interferon improves clinical outcomes Notwithstanding the “Background” section of the abstract, which says that the clinical benefits of treatment with interferon have not been established (implying that this question will be addressed in the current study), the authors did not compare clinical outcomes in treated versus nontreated groups They compared patients within these groups (i.e., in the treatment group, they compared patients with clearance of HBeAg to those without clearance.) They showed that, within the group treated with interferon, those with clearance of HbeAg better than those that never achieved clearance The clinical outcomes you can compare in the abstract are death and liver transplantation These occurred in 8/103 treated patients and 5/53 untreated patients – little suggestion of any benefit This study suggests that treatment with interferon alfa is associated with higher rates of clearance of HBeAg, but the design used to draw this inference (following two different convenience samples, no random allocation or blinding) is weak, and clearance of HbeAg is a surrogate outcome A key point is that, even if treatment allocation were randomized and were shown to increase clearing of HBeAg and even if clearing of HBeAg were shown to correlate with improved clinical outcome, we would not know whether treatment with interferon improves outcome The reason is that the outcome in those with clearing of HBeAg is not what is relevant The relevant outcome is what happens to the everyone who is treated Thus, you would want to see an improved outcome in the entire treated group compared with the entire control group The reason for this important point is that, unless you see improvement in the group as a whole, you can’t tell whether treatment simply sorts patients into those who would have done well anyway (those who clear virologically) and those who would have done poorly It’s our old friend “once randomized always analyzed” helping out again If you divide the subjects into those who and not respond to treatment and then compare outcomes among those who and not respond, the treatment is likely to look beneficial! 282 Answers to problems 4a The main outcome is (very) subjective: a ≥50% reduction of headache within 15 minutes 4b If (as seems likely), the lidocaine caused numbness of the nose, this would interfere with the blinding Lack of blinding is a particular problem when (as is the case here) the outcome is subjective (The problem would be worse if the subjects knew the study was comparing a local anesthetic to placebo If all they knew was that two different treatments were being compared it might not be as clear to them whether they were getting study drug vs placebo.) 4c We disagree with the conclusion The best estimate of the relief provided by intranasal lidocaine would be the difference between the lidocaine and placebo groups, which was 55% − 21% = 34%, not 55% (The point of doing a doubleblind trial is to compare treatment with placebo!) Also, ≥50% reduction is not quite the same as “relief,” and the probable lack of blinding may lead to inflation of the apparent benefit 5a 11.8% − 9.4% = 2.4% 5b NNT = 1/ARR = 1/2.4% ≈ 42 5c pills/person treated per day × $5/120 pills × 42 people need to treat × 30 days/cardiovascular death prevented = $104/cardiovascular death prevented 5d The RRR was 14%, so the RR would be − 14% = 0.86 The risk in the tPA group would be the risk in the SK group × the RR: 0.86 × 7.3% = 6.3% 5e ARR = 7.3% − 6.3% = 1.0% (Alternatively, could also 14% × 7.3% = 1.0%.) 5f The NNT = 1/ARR = 1/1% = 100 5g Additional cost of tPA = $3400 − $560 = $2840 NNT = 100 NNT × Cost = 100 × $2840 = $284,000 per death prevented (Aspirin is a better deal!) 6a RR = (1 − 40.2%)/(1 − 26.7%) = 59.8%/73.3% = 0.816 RRR = − RR = 0.184 = 18.4% ARR = 73.3% − 59.8% = 13.5% 6b 1/ARR = 1/.135 = 7.4 6c The cost per pill is $180/60 = $3, and treatment twice a day for a week requires 14 pills, so the cost per week is 14 × $3 = $42 Since 7.4 patients need to be treated for each one that responds, the cost per responding patient is 7.4 × $42, about $311 6d If each responder costs $311/week and has more CSBMs per week, the cost per additional CSBM is about $311/2, or $155 Chapter 10 Problem answers: alternatives to randomized trials The greatest strength of causal inference would come from comparing autism rates in children of Rh− and Rh+ mothers before 2001 If there is any difference, it would be useful to determine whether it persists for infants conceived after thimerosal was removed from Rhogam 283 Chapter 10 Problem answers: alternatives to randomized trials 2a 2b 2c 4a 4b Note that you wouldn’t want just to compare the risk in offspring of mothers who received Rhogam to those who did not, because confounding factors, like reliable attendance at prenatal visits, might be associated with getting Rhogam and also with the diagnosis of autism (You could, however, make this comparison if you did it both before and after thimerosal was removed.) The least satisfactory comparison would be between everyone exposed to thimerosal and everyone not exposed, including both time periods, because the exposure to thimerosal (which varied over time) would be confounded by changes in diagnostic criteria (which also varied over time) The predictor variable should be screened/not screened and/or frequency of screening with DRE The denominators should be men at risk of prostate cancer who either were or were not screened Denominators could also be person-years of follow-up following screening or failure to screen The key point is that the denominator should not be restricted to men who develop prostate cancer Possible ways of controlling or evaluating confounding/selection bias are: i) Try to control confounding by multivariate adjustment by age, number of health maintenance visits, smoking, family history, race, etc if data are available ii) Try to control confounding using propensity score analysis: look at other predictors in the dataset of receiving DRE, then create a propensity score for DRE and stratify, match, or control for it in multivariate analyses iii) To assess the likelihood of confounding, look at other predictor variables that might be affected by volunteer bias (e.g., number of measurements of serum cholesterol) and see if they are associated as strongly as DRE with decreased deaths from prostate cancer If so, the apparent benefit of DRE is likely to be due to confounding or selection bias, especially if it is diminished after adjusting as suggested under (i) and (ii) above iv) Alternatively, you could assess the likelihood of confounding by examining whether DRE also appears to protect against other outcomes one might expect to be affected by selection bias – that is, see if DRE is associated with decreased deaths from other causes, like heart disease and lung cancer If such nonspecific benefits are found, confounding or selection bias is likely The question is whether it is intermittent ibuprofen use itself, or something associated with it (e.g., headaches) that might alter the risk of colon cancer Asking about acetaminophen use acts as a control exposure If it, too, is associated with reduced colon cancer risk, confounding by indication would be a greater concern Confounding by indication could make the vaccine appear less effective than it really is This would occur if older, sicker people (i.e., the ones most likely to die during the flu season and therefore the group in whom the vaccine was most indicated) were over-represented in the vaccinated group Volunteer bias could make the vaccine look falsely good This would occur if patients in better health or with better health habits or access to care were over-represented in the vaccinated group 284 Answers to problems 4c The answer to this problem is analogous to the example of looking at deaths due to colon cancers beyond the reach of the sigmoidoscope in the study of sigmoidoscopy discussed in the chapter The investigators could see if the flu vaccination was associated with reduced mortality during seasons other than the flu season (i.e., the summer months) Because we specifically refer to unmeasured and unmeasurable confounders, propensity scores and other multivariate techniques won’t work for this problem 5a The propensity score for each subject in the study was the predicted probability (from a multivariable model) that he or she would be treated perioperatively with lipid-lowering agents This is to control for confounders that affect both the likelihood of receiving therapy and mortality risk 5b i) The left-most column is the mortality for people at lowest probability of receiving lipid-lowering therapy, who nonetheless did receive it, so there are not very many of them In fact, the legend to the figure tells you that only 0.5% of 156,114 (781 people) in that quintile were so treated! This leads to the wider confidence interval, reflected by that error bar ii) The suggestion that people with the lowest propensity for treatment might be harmed should make you cautious about promoting perioperative lipidlowering treatment in all patients not currently receiving it The result suggests that perhaps people prescribing these medicines actually know some things that are not captured in the model, that allow them only infrequently to give medication to people who are not appear to benefit However, based on the footnote of the table, since even subjects in the highest propensity quintile had low (∼31%) use of these drugs, if the results are real and causal, there will still be plenty of people not getting the drugs now who might benefit from them Chapter 11 Problem answers: understanding P-values and confidence intervals 1a False, P-values are conditional on the null hypothesis being true 1b False, the null hypothesis usually states that there is no difference between groups, so with sufficiently convincing data, you can reject the null hypothesis and conclude that there is a difference 1c True, high P-values are consistent with (but not prove) the null hypothesis 1d False, although, if you did the study 100 times, you would expect that, in 95% of them, the 95% CI for the study would include the true value; once the study is completed, other information must also be considered It is correct that an abnormal test ordered as part of a 20-test panel is more likely to be a false-positive than when the test is ordered by itself, but only because it is likely to have a lower prior probability Given the same clinical situation (i.e., the same prior probability), it makes no difference how many other tests you order at the same time 285 Chapter 11 Problem answers: understanding P-values and confidence intervals 3a Since the numerator was 2, the upper limit of the 95% CI will be about 7/259 = 2.7% (The exact upper limit is 2.76%.) 3b The upper limit of the 95% CI for the risk difference is only a 0.5% increase in total mortality – well below the 2% increase felt to be clinically significant by the editorialists What seems to be an underpowered study may not be underpowered if the goal was to rule-out significant harm and the trend is toward benefit (Similar conclusions apply to the adverse events other than death.) 3c They might have trouble believing the results if their estimate of the prior probability of lower mortality in the sentinel-node group was very low 4a Given the incredibly wide confidence interval, the study provides very little information on this hypothesis Therefore, all you can say about the posterior probability is that it is probably not much different from the prior probability 4b The sample size was probably very small (This is probably because UTIs were uncommon among the women who had intercourse less than once a week, and may also be because both diaphragm use or oral contraceptive use were uncommon in this group of women who infrequently needed contraception.) 5a Because the Bonferroni correction does not take prior probability into account and is overly conservative, we hardly ever think it is appropriate In this case, it is definitely too conservative: the prior probability that an active drug will cause more adverse effects than placebo is never very low, and in the case of adverse psychiatric effects of a psychiatric drug, seems particularly high This is a good example of why not to use Bonferroni! 5b We disagree What the treating physicians thought in this double-blind study is irrelevant If treating physicians could determine causality, there would be no need to randomized, double-blind trials The strength of evidence for causality is based on the magnitude of the excess of events in the treated group compared with the placebo group and the likelihood of alternative explanations for that excess (In a properly randomized and blinded trial, the only alternative explanation to causality is chance.) The Bonferroni correction is a conservative correction that makes it harder to reject the null hypothesis This is like requiring a test to be more abnormal before calling it positive It will tend to decrease both true-positive and false-positive results, decreasing sensitivity and increasing specificity Index AAA See abdominal aortic aneurysm abdominal aortic aneurysm (AAA), reliability of testing for, 37–38, 261 with screening tests, 135–136, 273–274 absolute risk increase (ARI), 198 absolute risk reduction (ARR), 198 confidence intervals and, 231 ACI-TIPI See Acute Coronary Ischemia – Time Insensitive Predictive Instrument Acquired Immune Deficiency Syndrome (AIDS), dichotomous tests for, 66, 263 acute coronary ischemia, diagnosing of, Acute Coronary Ischemia – Time Insensitive Predictive Instrument (ACI-TIPI), 175 acute myocardial infraction (AMI), randomized trials for, 203–204, 282 adjustment from anchor, in probability, 245–246 alternative diagnosis, 227 alternatives to randomized trials for autism factors, 217, 282–283 for colon cancer, 217–218, 283 for flu vaccines, 218, 283–284 for prostate cancer, 217, 282–283 American Cancer Society, 240 American College of Obstetricians, 240 American College of Radiology, 240 AMI See acute myocardial infraction, randomized trials for anchoring bias, 246–247 antidepressants randomized trials with, confidence intervals in, 237–238, 285 appendicitis double gold standard bias and, 115, 273 in multiple tests, 180–181, 277–278 reliability of testing for, 34, 256–257 area under an ROC curve (AUROC), 70–72 discrimination in prognostic tests and, 143 ARI See absolute risk increase ARR See absolute risk reduction as-treated analysis, 189–190 asthma, screening tests for, 135, 273 AUROC See area under an ROC curve 287 autism, randomized trials for causes of, 217, 282–283 availability, in probability, 244–245 axillary dissection, 235–236 B See cost of failing to treat bacteremia, 6, 45, 69, 71, 73–74, 79–80, 82–84, 87, 165, 167–168, 177, 225, 229, 231 continuous/multilevel testing for, 91–92, 268–269 bacterial meningitis continuous/multilevel testing for, 91–92, 268–269 randomized trials for, 201–202, 280 bias See also double gold standard bias; spectrum bias; verification bias anchoring, 246–247 cognitive, 246 confirmation, 246 definition of, 244 in diagnostic test studies, 99–107 application of, 109 double gold standard, 101–103 incorporation, 100 overfitting, 106–107 spectrum, 102–106 verification, 100–101 laboratory error and, 227 lead time, 126 length time, 126–127 pseudodisease, 128–129 publication, 148 random error v., 10 result distortion from, 132 in randomized trials, 191 in screening tests, 125–129 slippery linkage, 130–131 spectrum, 102–106 definition of, 102–104 disease prevalence and, 104–105 ESR and, 104 in multiple tests, 162 sensitivity in, 102 specificity in, 102 test in-dependence and, 105–106 for UTIs, 105–106 stage migration, 127–128 sticky diagnosis, 130 288 Index bias (cont.) verification, 99–101 volunteer, 125–126 binding, 187 biopsies, kidney, Bland-Altman plot, 10 variable types and, for reliability, 11 blinding, in prognostic tests, 147 blinding, in randomized trials, 187 blue belts, 231 bone cancers, prognostic tests for, 153–154, 276–277 Boneferroni correction, 226 confidence intervals and, 238, 285 breast cancer, 6, 44–45, 51, 118, 128, 131–132, 144, 149, 192–193, 240, 246 brown belt, 231 Browner, Warren, 224 C See cost of treating nondisease calibration, 31 for low back pain, 141–143 in prognostic tests, 140 cancer screening tests, 101–102 in EBM, 240–241 prognostic, 151–152, 275 Cat Scratch Fever, dichotomous tests for, 65, 262 cardiac arrhythmia suppression trial, 118 categorical variables, 11 inter-observer agreement for, 11–12, 13, 15, 17, 19, 21 CHD.See congenital heart disease, screening tests for chest pain (CP), reliability of testing for, 33–34, 256 CHF See congestive heart failure, diagnostic test studies for chronic hepatitis, randomized trials for, 202–203, 281 chronic obstructive pulmonary disease (COPD), continuous/multilevel testing for, 90–91, 267 clinical decision rules, 175–177 coefficient of variation, and reliability, 10 cognitive bias, 246 colic, testing for, 8–9, 255 colon cancer screening tests for, 134, 273 treatment effects on, 217–218, 283 composite endpoints, 188 computed tomography (CT), dichotomous tests for positive scans, 65–66, 262–263 likelihood ratios for, in minor head injuries, 66 conclusions, in diagnostic test studies, 97–98 confidence intervals, 222–229, 230–233, 234, 236, 238 in antidepressant randomized trials, 237–238, 285 Boneferroni correction and, 238, 285 power v., 232 P-values and, 229–232 sentinel-node biopsy v axillary dissection, 235–236 for small numerators, 233 with UTI studies, 237, 285 confirmation bias, 246 confounding by indication, 206–207 suppression in, 207 congenital heart disease (CHD), screening tests for, 136–137, 275 congestive heart failure (CHF), diagnostic test studies for, 114–115, 272 congenital hypothyroidism, consecutive sample, 101 constipation, randomized trials for, 204, 282 continuous measurements, reliability in, 23–28, 29 correlation coefficients and, 25–26 error by magnitude with, 26–28 in test-retest, 23 within-subject standard deviation/repeatability, 23–24 continuous test, 68–85, 88–93 See also receiver operating characteristic curve ROC curves in, 70–78 for WBC count, 68–69 logistic regression for, 173–175 continuous testing for bacteremia, 91–92, 268–269 for bacterial meningitis, 91–92, 268–269 for COPD, 90–91, 267 for Down syndrome, 92–93, 269–270 for home pregnancy tests, 88–89, 264–265 in hypothetical trial cases, 91, 267–268 for myeloperoxidase, 89, 265–266 continuous variables, 11 COPD See chronic obstructive pulmonary disease, continuous/multilevel testing for cost of failing to treat disease (B), 53, 57, 59, 61, 63, 177 cost of test (T), 57–60, 61, 63 cost of treating nondisease (C), 59, 61, 63, 177 CP See chest pain, reliability of testing for CT See computed tomography custom weights, 20–21 cytomegalovirus (CMV), prognostic tests for, 152, 275 D+/D− See dichotomous disease state decision problems, in diagnoses, 5–6 decision rules, 106, 163, 174–179, 249–250 Decision rules, multivariable, 156–185 derivation sets, 178–179 diagnosis, in EBM, of acute coronary ischemia, alternative, 227 with CT, D+/D− and, 4–5 decision problems in, 5–6 purpose of, 1–3 testing in, 4–5 accuracy of, reliability of, usefulness of, value of, 7–8 diagnostic test, 4–5.See alsobias; double gold standard bias; spectrum bias; verification bias accuracy of, for CHF, studies for, 114–115, 272 for colic, 8–9, 255 dipstick urine testing v urine microscopy and, studies for, 112, 271 for ectopic pregnancy, studies for, 112–113, 271 for intussusception, studies for, 113–114, 272 for metastatic undifferentiated carcinoma, 9, 255–256 prognostic v., 138–139 P-values and, analogies with, 224, 225 reliability of, 289 Index for rotavirus, for gastroenteritis, 8, 255 for sexual abuse, in prepubertal girls, 35, 258–259 studies for, 94–109 appraisal of, 95 biases for, 99–107 conclusions in, 97–98 design for, 94–96, 98 outcome variables, 97 predictor variables, 97, 98 research questions, 94, 98 results in, 97, 98 step-by-step appraisal of, 98 subjects in, 96–97, 98 systematic reviews of, 107–109 ROC curve in, 108 usefulness of, value of, 7–8 for vWD, studies for, 111, 270 dichotomous disease state (D+/D−), 4–5 dichotomous tests, 39–60 accuracy in, 41 benefit/cost quantification in, 52–55 for Cat Scratch Fever, 65, 262 definitions of, 42, 61 for enplanements, 67, 263 for Grunderschnauzer disease, 65, 262 for HIV/AIDS, 66, 263 information combinations within, 43 likelihood ratios for, 45–47, 49, 51 derivation of, 49–51, 62 multiple, 159–163 logistic regression, 169–171 result combination, 161, 162–163 spectrum bias, 162 negative predictive value and, 40–41 for positive CT scans, 65–66, 262–263 positive predictive value in, 40 prevalence in, 40 sampling schemes for, 41–42 sensitivity, 39–40 specificity, 40 for streptococcal infection, 65, 262 testing thresholds for, 52, 53, 55, 57, 59 treatment thresholds for, 52, 53–55, 57, 59 expected costs of, 53–54 no treat-test, 55 testing of, 55–56 test-treat, 55 2×2 table methods in, 41–46, 49 blank, 44 completed, 45 for influenza tests, 40, 41–49 dipstick urine testing, diagnostic testing studies for, 112, 271 discrete variables, 11 discrimination, in prognostic tests, 141–142 AUROC and, 143 for low back pain, 141–143 diseases acute coronary ischemia, classification systems for, 2–3 definition of, 3–4 heterogeneous, 158 pre-symptomatic, probability estimates for, 248–249 screening tests for, 117–119 double gold standard bias, 101–103 for appendicitis, in diagnostic testing of, 115, 273 in cancer screening tests, 101–102 numerical examples of, 103 sensitivity/specificity distortion in, 102 Down syndrome, continuous/multilevel testing for, 92–93, 269–270 EBM See evidence-based medicine ectopic pregnancy, diagnostic testing studies for, 112–113, 271 enplanements, dichotomous tests for, 67, 263 equivalency trials, 191 erythrocyte sedimentation rate (ESR), 104 likelihood ratios for, 104 ESR See erythrocyte sedimentation rate evidence-based medicine (EBM), barriers to idealized process of, 243–250 clinicians as, 243–249 cognitive errors as, 243–249 oversimplification of diagnostic issues as, 249–250 probability as, 243–249 cancer screening tests in, 240–241 criticisms of, 239–243 limits to clinician autonomy as, 241–243 nihilism of, as basis for, 240–242 randomized trials and, 239–240 definition of, diagnosis in, future applications of, 251 as malpractice, 242 in media, 240–241, 242 practicality of, 249–250 probability in, 243–249 post-test estimate errors in, 246–249 pre-test estimate errors in, 243–244 PSA in, 242 treatment in, disease classification systems and, 2–3 expected agreements, with kappa statistic, 13–14 balanced/unbalanced, 15–16 calculations of, 13 definition of, 13–14 false-negatives, P-values and, 222–224, 225 false-positives, P-values and, 222–224, 225 fecal occult blood screening, 132 flu vaccines, treatment effects on, 218, 283–284 focal segmental glomerulosclerosis, Framingham Risk Score (FRS), prognostic tests and, 152–153, 276 FRS See Framingham Risk Score, prognostic tests and gastroenteritis, diagnostic testing for, 8, 255 genetic tests, 148–150 evaluation/interpretation of, 149–150 Glasgow Coma Scale, 11 reliability of testing for, 35–37, 259–261 green belts, 230 Grunderschnauzer disease, dichotomous tests for, 65, 262 GUSTO study, 204 Guyatt, Gordon, hazard ratios, 140, 144 Health Plan Employer Data and Information Set (HEDIS), 121 290 Index Health Professionals study, 212 Healy, Bernadine, 240, 242 HEDIS See Health Plan Employer Data and Information Set heterogeneous disease, 158 heterogeneous nondisease, 158 HIV See human immunodeficiency virus home pregnancy tests, continuous/multilevel testing for, 88–89, 264–265 human immunodeficiency virus (HIV) dichotomous tests for, 66, 263 incorporation bias, 100 independence, of tests, 156–158 definition of, 156–157 non-independence v., 157–158 heterogeneous disease and, 158 heterogeneous nondisease and, 158 likelihood ratio slide rule and, 158–159 similarity of measurements in, 157–158 probability and, 157 index testing, 99 influenza testing treatment thresholds for, 60 2×2 table methods for, 40, 41–49 instrumental variables, 207–210 intentionally ordered tests, 225–226 intention-to-treat analysis, 189–190, 209 International Reflux Study in children, 232 inter-rater reliability, 10 See also kappa statistic kappa statistic for, classifications of, 22 expected agreements with, 13–14 formula for, 13, 14–15 good, 22 for three or more categories, 16–22 measures of, 10 intussusception, diagnostic studies for, 113–114, 272 kappa statistic, 11, 12–22 classifications of, 22 expected agreements with, 13–14 balanced/unbalanced, 15–16 calculations of, 13 definition of, 13–14 formula for, 13, 14–15 good, 22 for three or more categories, 16–22 with custom weights, 20–21 with linear weights, 17–18, 20 with quadratic weights, 21–22 unweighted, 17 linear weights, 17–18, 20 for four categories, 18 Kawasaki disease multiple tests for, 181–182, 278–279 kidney biopsies, laboratory error, bias and, 227 lead time bias, 126 in lung cancer screening tests, 135, 273 in nephritis screening tests, 134, 273 length time bias, 126–127 in lung cancer screening tests, 135, 273 in posterior fossa medulloblastoma screening tests, 136, 275 letrozole, 192–193, 195 likelihood ratios, 45–47, 49, 51 in CT tests, 66 definitions of, 49 derivation of, 49–51, 62 for ESR, 104 for multilevel tests, 79–81 interval, 79–81 odds ratios v., 170–171 recursive partitioning and, 164–165 slide rule for, 51, 86 for non-independent tests, 158–159 in testing threshold visualization, 56 for WBC count, 79 lipid-lowering agents, treatment effects on, 218, 284 logarithms, 86–87 logistic regression clinical decision rules with, cost in, 176–177 test selection in, 178 modeling for, 171–172 with ACI-TIPI, 175 Ottawa Ankle Rule in, 177 with PORT Pneumonia Score, 175, 176 in multiple tests, 168–177 dichotomous, 169–171 odds ratios and, 169–171, 172 for single continuous test, 173–175 low back pain, 141–143 low back pain, prognostic tests for, 152, 275–276 lung cancer, screening tests for, 135, 273 malpractice, EBM as, 242 mammography, 6, 44, 60, 120–122, 130–132, 240, 242, 247 Mayo Lung Study, 129 Menger, Fred, 106 metastatic undifferentiated carcinoma, testing for, 9, 255–256 migraine headaches, randomized trials for, 203, 282 minimal change disease, multilevel testing, 68–85, 88–93 for bacteremia, 91–92, 268–269 for bacterial meningitis, 91–92, 268–269 for COPD, 90–91, 267 for Down syndrome, 92–93, 269–270 for home pregnancy tests, 88–89, 264–265 in hypothetical trial cases, 91, 267–268 likelihood ratios for, 79–81 for myeloperoxidase, 89, 265–266 optimal cutoffs for, 81–82, 83 graphical approach to, 82–84 ROC curves and, 85 probability for, 81 multiple hypotheses Bonferroni correction and, 226 multiple tests and, 226–227 multiple tests, 156–179 See also independence, of tests; logistic regression for appendicitis, 180–181, 277–278 derivation sets and, 178–179 dichotomous, 159–163 result combination of, 161, 162–163 spectrum bias in, 162 independence in, 156–158 definition of, 156–157 non-independence v., 157–158 probability and, 157 291 Index for Kawasaki disease, 181–182, 278–279 logistic regression in, 168–177 clinical decision rules with, 175–177 dichotomous, 169–171 modeling for, 171–172 odds ratios and, 169–171, 172 for single continuous test, 173–175 for MI, 184, 279 multiple hypotheses and, 226–227 recursive partitioning in, 163–168 likelihood ratios and, 164–165 Ottawa Ankle Rule and, 163, 165 PROS Febrile Infant Study and, 165 software for, 166–167 treat fewer rule and, 183–184, 279 treat more rule and, 183–184, 279 validation sets and, 178–179 for WBC counts, 183–184, 278–279 myeloperoxidase, continuous/multilevel testing for, 89, 265–266 myocardial infarction AMI, randomized trials for, 203–204, 282 multiple tests for, 184, 279 nasal bone, 6, 159–166, 169, 171–172, 178 negative predictive value, 40–41 nephritis, screening tests for, 134, 273 lead time bias in, 134, 273 nephrotic syndrome, New England Journal of Medicine, 90 nominal variables, 11, 68 non-independence, of tests, 157–158 heterogeneous disease and, 158 heterogeneous nondisease and, 158 likelihood ratio slide rule and, 158–159 similarity of measurements in, 157–158 no treat-test threshold, 55 nuchal translucency, 92–93, 160 null hypothesis, 220–221 number needed to harm, 198–199 number needed to treat, 117, 192–195, 197, 231 Nurses’ Health study, 212 odds ratios, 140 likelihood ratios v., 170–171 in logistic regression, for multiple tests, 169–171, 172 in randomized trials, 194 OME Seeotitis media with effusion, randomized trials for optimal cutoffs, in multilevel testing, 81–82, 83 graphical approach to, 82–84 ROC curves and, 85 ordinal variables, 11, 68 continuous, 68 discrete, 68 Glasgow Coma Scale as, 11 otitis media with effusion (OME), randomized trials for, 201, 280 Ottawa Ankle Rule, 163, 165, 177 in logistic regression modeling, 177 outcome variables, 97, 98 multiple/composite, 147 of prognostic tests, 138 in randomized trials, 188, 191–192 additional measurements for, 211–212 dichotomous, 192–194 overfitting, 147 overfitting bias, 106–107 partitioning, recursive, 163–168 PCR tests Seepolymerase chain reaction tests Pediatric Research in Office Settings (PROS) Febrile Infant Study, 165 per-protocol analysis, 189–190 PID See positive in disease polymerase chain reaction (PCR) tests, 39 PORT Pneumonia Score, 175 calculation of, 176 positive in disease (PID), 39 positive predictive value, 40 posterior fossa medulloblastoma, screening tests for, 136, 274–275 length time bias in, 275 power, confidence intervals v., 232 predictive value, 40–41 predictor measurement, 212 predictor variables, 97, 98 Pre-symptomatic diseases, prevalence, in dichotomous tests, 40 probability, 47–48 See also treatment threshold probability adjustment from anchor in, 245–246 availability in, 244–245 diseases and, estimates for, 248–249 in EBM, 243–249 post-test estimate errors in, 246–249 pre-test estimate errors in, 243–244 likelihood ratios and, 45–47, 49, 51 in CT tests, 66 definition of, 49 derivation of, 49–51 slide rule for, 51 for multilevel testing, 81 negative predictive value and, 40–41 positive predictive value and, 40 posterior probability, 43–46, 49–51, 53, 55–57, 60–62, 80–81, 83, 94, 118, 145, 156–157, 170–171, 181, 224–225, 227–229 predictive value, 39–43, 45, 90, 96, 111–114, 222–223 prior probability, 7, 43–46, 49, 51–52, 55–58, 60–62, 66, 80–81, 86–87, 89, 92, 94, 97, 105, 116, 118, 156–157, 165, 171, 223–229, 241, 244 propensity scores, 213 P-values and, 222–224, 225 representativeness in, 244 specificity and, 40 test independence and, 157 treatment thresholds and, 52, 53–55, 57, 59 expected cost of, 53–54 formulas for, 62–64 no treat-test, 55 testing of, 55–56 test-treat, 55 2×2 table methods and, 41–46, 49 blank, 44 completed, 45 prognosis, 138 prognostic tests, 138–150 accuracy of, calibration in, 140 292 Index prognostic tests (cont.) discrimination in, 141–142 quantification of, 140–143 blinding in, 147 for bone cancers, 153–154, 276–277 for cancer, 151–152, 275 for CMV, 152, 275 diagnostic v., 138–139 effects of treatment in, 146 follow-up losses in, 146–147 FRS and, 152–153, 276 genetic, 148–150 evaluation/interpretation of, 149–150 hazard ratios in, 140, 144 for low back pain, 152, 275–276 multiple/composite outcomes in, 147 new information quantification in, 148 outcome variables of, 138 overfitting in, 147 publication bias in, 148 rate ratios in, 144 risk factors v., 139–140 risk ratios in, 140, 144 sample selection in, 146 sample size in, 147–148 for TP53 gene mutations, 154, 277 value assessment for, 144–145 propensity scores, 212–216 analysis of, 215–216 probability in, 213 PROS Febrile Infant Study SeePediatric Research in Office Settings Febrile Infant Study prostate cancer, treatment effects on, 217, 282–283 prostate-specific antigen (PSA), in EBM, 242 PSA See prostate-specific antigen pseudodisease, 128–129 See also double gold standard bias in Mayo Lung Study, 129 types of, 129 publication bias, 148 P-values, 222–233, 234, 236, 238 as conditional probability, 222–224, 225 confidence intervals and, 229–232 ARR and, 231 blue belt, 231 brown belt, 231 green belt, 230 white belt, 230 yellow belt, 230 definition of, 221–222 diagnostic test analogies with, 224, 225 false-positive/false-negative confusion with, 222–224, 225 intentionally ordered tests and, 225–226 quadratic weights, 21–22 random error, bias v., 10 randomized trials additional outcome measurements in, 211–212 for AMI, 203–204, 282 analysis of, 189–191 as-treated, 189–190 intention-to-treat, 189–190, 209 multiple comparisons in, 190–191 per-protocol, 189–190 subgroup, 190 between v within-group comparisons in, 191 for bacterial meningitis, 201–202, 280 bias directions in, 191 binding in, 187 for chronic constipation, 204, 282 for chronic hepatitis, 202–203, 281 composite endpoints in, 188 conduct of, 187–189 confidence intervals, with antidepressants, 237–238, 285 confounding by indication in, 206–207 suppression in, 207 critical appraisal of, 187, 189 design of, 187–189 for EBM, 239–240 effect size measurement in, 192 equivalency trials and, 191 follow-up losses in, 189 GUSTO study, 204 Health Professionals study, 212 instrumental variables v., 207–210 for migraine headaches, 203, 282 number needed to harm and, 198–199 Nurses’ Health study, 212 for OME, 201, 280 predictor measurement in, 212 propensity scores v., 212–216 analysis of, 215–216 probability in, 213 purpose of, 186–187 of screening tests, 124–125, 130–131 total v cause-specific mortality in, 130–131 set-up for, 186 surrogate outcomes in, 188 treatment cost in, 194–198 bad outcome v traditional, 196 uncertainty about patient disease and, 197–198 treatment effect quantification with, 186–199 alternatives to, 206–216 continuous outcome variables in, 191–192 dichotomous outcome variables in, 192–194 effect size inflation in, with odds ratio, 194 relative v absolute measures of, 192–193 unrelated variable measurements in, 211 rate ratios, 144 ratios See hazard ratios; likelihood ratios; odds ratios; rate ratios; risk ratios receiver operating characteristic (ROC) curve, 70–78 approach to, 74–78 AUROC and, 70–72 in diagnostic testing, systematic reviews of, 108 information in, 78 optimal cutoffs and, 85 in signal detection theory, 70 SROC, 108–109 WBC counts and, 74 Wilcoxon Rank Sum test and, 77 recursive partitioning, 163–168 likelihood ratios and, 164–165 Ottawa Ankle Rule and, 163, 165 PROS Febrile Infant Study and, 165 software for, 166–167 reliability, in testing, 10–32 See also kappa statistic; variable types, reliability and for AAA, 37–38, 261 for appendicitis, 34, 256–257 293 Index bias/random error, 10 Bland-Altman plot for, 10 variable types and, 11 coefficient of variation and, 10 of continuous measurements, 23–28, 29 average standard deviations and, 24 correlation coefficients and, 25–26 error by magnitude with, 26–28 in test-retest, 23 within-subject standard deviation/repeatedly, 23–24 for CP, 33–34, 256 for diagnosing sexual abuse, in prepubertal girls, 35, 258–259 for Glasgow Coma Scale scores, 35–37, 259–261 inter-rater, 10 kappa statistic for, 11, 12–22 measures of, 10 intra-rater, 10 literature studies of, 31–32 method comparison for, 28–31 calibration for, 31 standard deviation and, 10 variable types and, 11 Bland-Altman plot and, 11 categorical, 11 continuous, 11 discrete, 11 kappa statistic and, 12–22 nominal, 11, 68 ordinal, 11, 68 representativeness, in probability, 244 research questions, in diagnostic test studies, 94, 98 results, in diagnostic test studies, 97, 98 risk ratios, 140, 144 ROC curve Seereceiver operating characteristic curve rotavirus testing, for gastroenteritis, 8, 255 sample selections consecutive, 101 in prognostic tests, 146 selective, 101 sample size, in prognostic tests, 147–148 San Francisco Chronicle, 193 screening tests, 116–132 See also pseudodisease; randomized trials, of screening tests for AAA, 135–136, 273–274 for asthma, 134, 273 biases in, lead time, 126 length time, 126–127 pseudodisease, 128–129 stage migration, 127–128 volunteer, 125–126 cardiac arrhythmia suppression trial, 118 for colon cancer, 134, 273 definitions of, 116–117 excessive, 119–122, 123 HEDIS and, 121 pressure, sources for, 119–121 false results from, 121–123 fecal occult blood, 132 goals of, 117 harms from, 119, 120–121 for lung cancer, 135, 273 for nephritis, 134, 273 for posterior fossa medulloblastoma, 136, 274–275 public support for, 121 randomized trials of, 124–125, 130–131 total v cause-specific mortality in, 130–131 studies of, 123–129, 131 appraisals of, 123–125, 127, 129, 131 observational, 125–129 types of, 117–119 for presymptomatic disease, 118 for risk factors, 118–119 for unrecognized symptomatic disease, 117–118 underutilization of, 123 selective sample, 101 sensitivity, 39–40 PID and, 39 sentinel-node biopsy, 235–236 sexual abuse in prepubertal girls, reliability of testing for, 35, 258–259 signal detection theory, 70 slide rule, for likelihood ratios, 51, 86 for nonindependent tests, 158–159 in testing threshold visualization, 56 slippery linkage bias, 130–131 small numerators, 233 specificity, 40 spectrum bias, 102–106 definition of, 102–104 disease prevalence and, 104–105 ESR and, 104 in multiple tests, 162 sensitivity in, 102 specificity in, 102 test nondependence and, 105–106 for UTIs, 105–106 SROC Seesummary receiver operating characteristic curve stage migration bias, 127–128 survival rates and, 128 standard deviation, reliability and, 10 statistical significance testing, 220–221 sticky diagnosis bias, 130 strep throat, 65 dichotomous tests for, 65, 262 study design, for diagnostic test studies, 94–96, 98 subgroup analysis, 190 subjects, in diagnostic test studies, 96–97, 98 summary receiver operating characteristic (SROC) curve, 108–109 suppression, 207 surrogate outcomes, 188 T See cost of test tests.See also continuous testing;diagnostic testing; dichotomous tests; multiple tests; prognostic tests; randomized trials; screening tests cancer screening, 101–102 continuous, 68–85, 88–93 dichotomous, 68–70 cost, T, 52 diagnostic, 4–5 accuracy of, prognostic v., 138–139 P-values and, analogies with, 224–225 reliability of, studies of, 94–109 294 Index tests (cont.) systematic reviews of, 107–109 usefulness of, value of, 7–8 dichotomous, 39–60 accuracy in, 41 benefit/cost quantification in, 52–55 continuous, 68–70 definitions of, 42, 61 information combinations within, 43 likelihood ratios for, 45–47, 49, 51 negative predictive value and, 40–41 PCR, 39 positive predictive value in, 40 prevalence in, 40 ROC curves in, 70–78 sampling schemes for, 41–42 sensitivity in, 39–40 specificity in, 40 testing thresholds for, 52, 53, 55, 57, 59 treatment thresholds for, 52, 53–55, 57, 59 2×2 table methods in, 41–46, 49 genetic, 148–150 evaluation/interpretation of, 149–150 index, 99 influenza, 40 treatment thresholds for, 60 2×2 table methods for, 41–49 intentionally ordered, 225–226 multilevel, 68–85, 88–93 likelihood ratios for, 79–81 optimal cutoffs for, 81–82, 83 probability for, 81 multiple, 156–179 derivation sets and, 178–179 dichotomous, 159–163 imaging finding outcomes in, 177–180 independence in, 156–158 logistic regression in, 168–177 recursive partitioning in, 163–168 validation sets and, 178–179 prognostic, 138–150 accuracy of, 139 blinding in, 147 diagnostic v., 138–139 effects of treatment in, 146 follow-up losses in, 146–147 genetic, 148–150 hazard ratios in, 140, 144 multiple/composite outcomes in, 147 new information quantification in, 148 outcome variables of, 138 overfitting in, 147 publication bias in, 148 rate ratios in, 144 risk factors v., 139–140 risk ratios in, 140, 144 sample selection in, 146 sample size in, 147–148 value assessment for, 144–145 reliability in, 10–32 screening, 116–132 biases in, 125–129 cardiac arrhythmia suppression trial, 118 definitions of, 116–117 excessive, 119–122, 123 false results from, 121–123 fecal occult blood, 132 goals of, 117 harms from, 119, 120–121 public support for, 121 randomized trials of, 124–125, 130–131 studies of, 123–129, 131 types of, 117–119 underutilization of, 123 statistical significance, 220–221 test-retest reliability, 23 test-treat threshold, 55 Time, 242 TP53 gene mutations, prognostic tests for, 154, 277 treat fewer rule, 183–184, 279 treat more rule, 183–184, 279 treatment, in EBM, disease classification systems and, 2–3 treatment threshold probability, 52, 53–55, 57, 59 expected cost of, 53–54 no treat-test, 55 testing of, 55–56 formulas for, 62–64 for imperfect test, 59–60 for influenza tests, 40, 60 likelihood ratio slide rule in, 56 for perfect test, 58–59 visualization of, 56 test-treat, 55 2×2 table methods, 41–46, 49 blank, 44 completed, 45 for influenza tests, 40, 41–49 trial cases, continuous/multilevel testing in, 91, 267–268 trisomy 21 See Down syndrome, continuous/multilevel testing for urinary tract infections (UTIs), spectrum bias and, 105–106 confidence intervals and, in studies of, 237, 285 VCUG for, 9, 256 urine microscopy, diagnostic testing studies for, 112, 271 U.S News and World Report, 240–241 U.S Preventative Health Services Task Force, 123 UTIs See urinary tract infections validation sets, 178–179 variable types, reliability and, 11 categorical, 11 inter-observer agreement for, 11–12, 13, 15, 17, 19, 21 continuous, 11 discrete, 11 instrumental, in randomized trials, 207–210 kappa statistic and, 12–22 nominal, 11, 68 ordinal, 11 outcome, 97, 98 of prognostic tests, 138 predictor, 97, 98 VCUG See voiding cystourethrogram, for UTIs verification bias, 100–101 voiding cystourethrogram (VCUG), for UTIs, 9, 256 volunteer bias, 125–126 295 Index von Willebrand disease (vWD), diagnostic testing studies for, 111, 270 vWD Seevon Willebrand disease, diagnostic testing studies for Walking man, approach to ROC curves, 74–78 WBC Seewhite blood cell counts, multiple testing for WBC count Seewhite blood cell count white belt, 230 white blood cell (WBC) count, 68–69 likelihood ratios for, 79 posterior odds of, 80 post-test odds of, 83 ROC curve and, 74 sensitivity/specificity of, 74 white blood cell (WBC) counts, multiple testing for, 183–184, 278–279 Wilcoxon Rank Sum test, 77 yellow belt, 230 ... Burlingame, California Evidence- Based Diagnosis Thomas B Newman University of California, San Francisco Michael A Kohn University of California, San Francisco CAMBRIDGE UNIVERSITY PRESS Cambridge, New... Singapore, São Paulo Cambridge University Press The Edinburgh Building, Cambridge CB2 8RU, UK Published in the United States of America by Cambridge University Press, New York www .cambridge. org Information... intentionally left blank Evidence- Based Diagnosis Evidence- Based Diagnosis is a textbook about diagnostic, screening, and prognostic tests in clinical medicine The authors’ approach is based on many years

Ngày đăng: 09/11/2019, 00:08

Từ khóa liên quan

Mục lục

  • Cover

  • Half-title

  • Title

  • Copyright

  • Contents

  • Preface

  • Acknowledgments & Dedication

  • Abbreviations/Acronyms

    • Greek letters

    • 1 Introduction: understanding diagnosis and diagnostic testing

      • Two areas of evidence-based medicine: diagnosis and treatment

      • The purpose of diagnosis

      • Definition of "disease"

      • Dichotomous disease state (D+/D-): a convenient oversimplification

      • Generic decision problem: examples

      • Preview of coming attractions: criteria for evaluating diagnostic tests

      • Summary of key points

      • References

      • Chapter 1 Problems: understanding diagnosis

      • References for problem set

      • 2 Reliability and measurement error

        • Introduction

        • Types of variables

Tài liệu cùng người dùng

Tài liệu liên quan