Patrick J. Bryant, Pharm.D.
, FSCIP
Director, Drug Information Center
Clinical Professor, Pharmacy Practice
University of Missouri–Kansas City School
of Pharmacy
Kansas City, Missouri
Heather A. Pace, Pharm.D.
Assistant Director, Drug Information Center
Clinical Assistant Professor, Pharmacy Practice
University of Missouri–Kansas City School
of Pharmacy
Kansas City, Missouri
American Society of Health-System Pharmacists®
Bethesda, Maryland
Any correspondence regarding this publication should be sent to the publisher, American Society of
Health-System Pharmacists, 7272 Wisconsin Avenue, Bethesda, MD 20814, attention: Special
Publishing.
The information presented herein reflects the opinions of the contributors and advisors. It should
not be interpreted as an official policy of ASHP or as an endorsement of any product.
Because of ongoing research and improvements in technology, the information and its applications
contained in this text are constantly evolving and are subject to the professional judgment and
interpretation of the practitioner due to the uniqueness of a clinical situation. The editors,
contributors, and ASHP have made reasonable efforts to ensure the accuracy and appropriateness
of the information presented in this document.
However, any user of this information is advised that the editors, contributors, advisors, and ASHP
are not responsible for the continued currency of the information, for any errors or omissions, and/
or for any consequences arising from the use of the information in the document in any and all
practice settings. Any reader of this document is cautioned that ASHP makes no representation,
guarantee, or warranty, express or implied, as to the accuracy and appropriateness of the
information contained in this document and specifically disclaims any liability to any party for the
accuracy and/or completeness of the material or for any damages arising out of the use or non-use
of any of the information contained in this document.
Director, Special Publishing: Jack Bruggeman
Acquisitions Editor: Hal Pollard
Senior Editorial Project Manager: Dana Battaglia
Project Editor: Johnna Hershey
Cover and Page Design/Composition: David A. Wade
Library of Congress Cataloging-in-Publication Data
Bryant, Patrick J.
The pharmacist's guide to evidence-based medicine for clinical decision
making / Patrick J. Bryant, Heather A. Pace.
p. ; cm.
Includes bibliographical references and index.
ISBN 978-1-58528-177-0
1. Pharmacy--Decision making. 2. Evidence-based medicine. I. Pace,
Heather A. II. American Society of Health-System Pharmacists. III. Title.
[DNLM: 1. Drug Therapy. 2. Decision Making. 3. Evidence-Based
Medicine. 4. Pharmacology, Clinical. WB 330 B915p 2008]
RS92.B765 2008
615'.1--dc22
2008034439
©2009, American Society of Health-System Pharmacists, Inc. All rights reserved.
No part of this publication may be reproduced or transmitted in any form or by any means,
electronic or mechanical, including photocopying, microfilming, and recording, or by any informa-
tion storage and retrieval system, without written permission from the American Society of Health-
System Pharmacists.
ASHP is a service mark of the American Society of Health-System Pharmacists, Inc.; registered in
the U.S. Patent and Trademark Office.
ISBN: 978-1-58528-177-0
ii
Table of Contents
Acknowledgments ....................................................................... iv
Preface ......................................................................................... vi
Contributors .............................................................................. viii
Chapter 1: Introduction ........................................................... 1
Chapter 2: Basics for Interpretation .................................... 11
Chapter 3: Step 1 – Define the Clinical Question .............. 41
Chapter 4: Step 2 – Retrieve Pertinent Information ........... 51
Chapter 5: Step 3 – Evaluate Literature .............................. 61
Chapter 6: Step 4 – Categorize Quality of Evidence .......... 87
Chapter 7: Step 5 – Develop a Conclusion and
Recommendation ............................................... 103
Chapter 8: Alternative Sources of Evidence ..................... 119
Chapter 9: Applications ....................................................... 127
Chapter 10: Applying Evidence-Based Medicine
Principles to Dietary Supplement
Therapeutic Decisions ...................................... 151
Appendix es
Appendixes
Appendix 1 –Glossary of Evidence-Based Medicine Terms
(including the Biostatistical Glossary) ........... 171
Appendix 2 –Evidence-Based Medicine Tool Kit .................. 187
Index ........................................................................................ 191
iii
Acknowledgments
T he development of a unique process like the one described
in this book is not an easy task and has involved many differ-
ent minds over several years. We would like to thank those resi-
dents and fellows that have trained in the University of Missouri–
Kansas City School of Pharmacy Drug Information Center for
their assistance and feedback used to modify and improve the
process.
These people include Antoine Richardson (also previous fac-
ulty), Jessica Beal, Michael Steinberg, Julie Kenkel, Kevin Clauson,
Kelly Shields, Marissa Curiel-Dickson, Elizabeth Poole, Celtina
Reinert, and Melita Croom. We would also like to thank those
drug information faculty who have made contributions to the
process, including Cydney McQueen (also previous Fellow),
Lindsey Schnabel (also previous Resident), Morgan Sperry, Karen
Norris, Angela Bedenbaugh, and Chris Meier.
Denise Woolf with our Center has been monumental in proof-
reading and suggesting changes that make the book more read-
able and understandable; we thank her for this contribution.
Over 600 students have passed through the evidence-based
medicine course and provided feedback to improve this process.
Specifically, we want to thank Amber Sawyer for reading through
the book and identifying evidence-based medicine terms for the
glossary from a student’s perspective.
Tim Candy, Manager, Global Medical/Clinical Affairs at Baxter
Healthcare Corporation, deserves a huge thank you for his con-
tributions to the Basics for Interpretation chapter. He was also
responsible for providing the biostatistics section of the Evidence-
Based Medicine Glossary. We also thank Seth Berry, Associate
Director, Modeling and Simulation at Quintiles, Inc., for his help
with the Basics for Interpretation chapter.
We would like to thank our Pharmacy Practice Faculty col-
leagues for their curb-side consults as we developed this process.
Specifically, we thank Brooke Patterson, Jennifer Santee, and
iv
Tatum Mead for reviewing the chapter on Basics for Interpreta-
tion and providing feedback on the clarity and ease of reading
this section of the book.
This project would never have happened without Hal Pol-
lard, Content Development & Acquisitions Editor at the Ameri-
can Society of Health-System Pharmacists (ASHP), who caught
and supported our vision for a “how to” book on this subject. We
thank you Hal for your continued encouragement and support
throughout this project. We would also like to thank Jack
Bruggeman, Director, ASHP’s Special Publishing, for providing
the resources to see this project through. We now have a better
appreciation for the job that Dana Battaglia, Senior Editorial
Project Manager at ASHP, does; she gets a special thank you for
training us to be book editors, not to mention the long hours she
spent reviewing and providing feedback for our multiple drafts.
Johnna Hershey, Director, ASHP’s Publications Production Cen-
ter in the Publications and Drug Information Systems Office, cer-
tainly deserves a big thank you for her copy editing and final
production work with this book. A thanks goes out to all those at
ASHP who have played some part in the development and pro-
duction of this book.
No project of this magnitude happens without the adminis-
trative support provided by the Chair of Pharmacy Practice,
Patricia Marken and Dean Robert Piepho. Thank you both for
your patience and guidance.
Perhaps most important is our thanks to each of our families
for their encouragement and patience while we took time away
from them to work on this project. They have played such a key
role in allowing this project to be completed.
Pat Bryant
Heather Pace
v
Preface
E vidence-based medicine has been taught in a focused manner
at the University of Missouri–Kansas City School of Phar-
macy since 1998. Over that time, a unique systematic approach
has been developed that caters to the analytical mind of the phar-
macy practitioner. This book represents an attempt to transfer
this process to pharmacy students and practitioners for maxi-
mizing clinical decision making.
Other evidence-based medicine processes have proven to be
complex, labor intensive, and time consuming. The 5-Step Evi-
dence-Based Process described in this book comes out of simplify-
ing the complexity while still maintaining adequate rigor. This
allows the practitioner to apply the process to every day, indi-
vidual patient and population-based, time sensitive decision
making.
This is a “how to” book not a text book, although we antici-
pate the book will be a valuable addition to literature evaluation,
evidence-based medicine, and drug information courses. The
book was especially developed for the busy practitioner who can
read through the text in an evening or two and immediately ap-
ply the 5-Step Evidence-Based Medicine Process to his or her prac-
tice setting. Although there is an assumed minimal level of phar-
macotherapy, literature evaluation, study design, and interpre-
tive biostatistics knowledge for the reader, reference to additional
reading is made to assist those requiring a review of these foun-
dational topics.
The book is arranged in three specific sections that include
an introduction and review of basic study interpretation skills,
the specific 5-Step Evidence-Based Medicine Process, and applica-
tion of this process to specific individual patient and population-
based situations. Think of the book as containing core knowl-
edge based on the 5-Step Evidence-Based Medicine Process (one
chapter for each step). In addition, chapters have been added
that will provide the required skills, resources, and examples to
put this process into practice.
vi
A seasoned practitioner may want to go directly to those chap-
ters describing the 5-Step Evidence-Based Process, referring to the
other chapters as needed. Other readers are encouraged to read
the book from cover to cover, keeping in mind that the book was
written and formatted with the intention to be an easy read.
Throughout the book, “Key Ideas” are extracted and set out from
the text. They serve as a quick review as well as reference points
to find specific sections of the text to refer back to as the process
is applied in clinical practice. In addition, figures and tables have
been used extensively to visually convey concepts. Examples have
been provided to illustrate application of these concepts. A Glos-
sary of Evidence-Based Medicine terms has been included at the
end of the book to help the reader become familiar with the unique
vocabulary associated with this discipline. An Evidence-Based
Medicine Tool Kit has also been provided to assist with the initial
application of this process to clinical practice.
Our goal is to provide this 5-Step Evidence-Based Medicine Pro-
cess to every pharmacy practitioner and student so they can
quickly learn and incorporate this process into their clinical de-
cision making. Our hope is that the application of this process to
clinical practice will result in overall improved patient care.
Pat Bryant
Heather Pace
vii
Contributors
Patrick J. Bryant, Pharm.D., FSCIP
Director, Drug Information Center
Clinical Professor, Pharmacy Practice
University of Missouri–Kansas City School of Pharmacy
Kansas City, Missouri
Timothy A. Candy, Pharm.D., M.S., BCPS
Manager, Global Medical/Clinical Affairs
Baxter Healthcare Corporation
Round Lake, Illinois
Cydney E. McQueen, Pharm.D.
Clinical Associate Professor, Pharmacy Practice
University of Missouri–Kansas City School of Pharmacy
Kansas City, Missouri
Heather A. Pace, Pharm.D.
Assistant Director, Drug Information Center
Clinical Assistant Professor, Pharmacy Practice
University of Missouri–Kansas City School of Pharmacy
Kansas City, Missouri
Celtina K. Reinert, Pharm.D.
Integrative Therapies Pharmacist
Sastun Center of Integrative Health Care
Overland Park, Kansas
viii
Antoine D. Richardson, Pharm.D.
Clinical Instructor
University of Missouri–Kansas City School of Pharmacy
Kansas City, Missouri
Lindsey N. Schnabel, Pharm.D.
Clinical Assistant Professor, Pharmacy Practice
Assistant Director, Drug Information Center
University of Missouri–Kansas City School of Pharmacy
Kansas City, Missouri
Morgan L. Sperry, Pharm.D.
Clinical Assistant Professor, Pharmacy Practice
Assistant Director, Drug Information Center
University of Missouri–Kansas City School of Pharmacy
Kansas City, Missouri
ix
Chapter 1
Introduction
Patrick J. Bryant
Evidence-Based Medicine
Sources of Knowledge
The Problem
The Solution
How This Book Will Help You
References
Evidence-Based Medicine
Evidence-based medicine focuses on the scientific method as the
key source of knowledge in making clinical decisions. Research
shows that when we use experience as the primary knowledge
source to make clinical decisions, we tend to overestimate effi-
cacy and underestimate risk factors of a specific drug or proce-
dure.1 This leads to variation in services and treatment, resulting
in inappropriate care, lack of care, and increase in health care
costs. An approach to making clinical decisions has emerged
within the medical discipline called evidence-based medicine.
Evidence-based medicine is an attempt to provide something other
than just experience of the practitioner in making clinical deci-
sions. David Sackett, one of the pioneers in this area, originally
coined the term “evidence-based medicine” while teaching medical
students; he essentially defined this term as:
“The conscientious, explicit, and judicious use of current best
evidence in making decisions about the care of individual pa-
tients, while integrating clinical experience with the best avail-
able evidence from a systematic search.”2
1
2 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Sources of Knowledge
As mentioned, evidence-based medicine utilizes the scientific
method as a key source of knowledge for clinical decision mak-
ing. However, in addition to the scientific method, there are four
other sources of knowledge.3 Each source or method presents
potential problems that are discussed below.
Key Evidence-based medicine focuses on
scientific method as the key source of
Idea knowledge to make clinical decisions.
1. Refer ence to tradition – accepting certain truths as giv-
Reference
ens. Problem: many traditions are not evaluated for va-
lidity nor tested against potentially superior alternatives.
2. Refer ence to authority – placing trust in those who are
Reference
authorities or experts on an issue. This can be useful where
scientific evidence is weak or unavailable. Problem: this
method minimizes the need for critical analysis and con-
firmation of validity and does not encourage testing of
potentially superior alternatives.
3. Trial and er
errror – applying multiple attempts to find a solu-
tion by chance. Used when no other basis for making a de-
cision exists. Problem: this method results in a haphazard
and unsystematic process to obtain knowledge that is gener-
ally not shared, limited in scope, and time consuming; it
also prevents identifying/confirming the best solution.
4. Logical rreasoning
easoning – involving deductive reasoning—a sys-
tematic method for drawing conclusions by using a se-
ries of three interrelated statements. Problem: deductive
reasoning only produces a hypothesis that still requires
testing since usefulness is dependent upon the truth of
the premises developed. Example
Example—all living things must
die (major premise), humans are living things (minor
Chapter 11: Introduction 3
premise); therefore, all humans must die (conclusion).
Logical reasoning also involves inductive reasoning or de-
veloping generalizations from specific observations. Prob-
lem: quality of the knowledge derived from inductive rea-
soning is dependent upon how well specific observations
represent the general situation. To be absolutely certain
of the conclusion, one must observe all possible examples
of the event, and that is rarely possible. Example
Example—edema
decreases with application of ice to an injured ankle. Af-
ter numerous observations of this phenomenon, one con-
cludes cold reduces fluid infiltration in body tissues.
5. Scientific method – applying a logical sequential pro-
cess to develop a conclusion. This process involves iden-
tifying the problem, organizing collection of data, objec-
tively analyzing the data, and interpreting the findings.
The goal is to enable other researchers to reproduce the
results that confirm validity. This is the most rigorous
process for obtaining new knowledge. Problem: complex-
ity and variability of components (e.g., nature/environ-
ment, unique psychosocial and physiological capacities
of individuals) introduce uncertainty into interpretation
and generalization of data.
The Problem
Although the best evidence comes from the scientific method,
medical practice continues to focus on the other four sources of
knowledge. The result is variation in medical practice patterns and
variation in treatment for virtually the same patient with the same
disease state.4 In addition, a gap exists between new research find-
ings and incorporating these findings into clinical practice.
Key Although the best evidence comes from the
scientific method, medical practice continues to
Idea focus on less accurate sources of knowledge.
4 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
As a result, the most recent scientifically developed knowl-
edge is not being applied to clinical practice. The scientific
method, our best source of knowledge, is not being maximized
to ensure the best patient care.
Several evidence-based medicine processes have been devel-
oped; however, these processes tend to be complex, labor inten-
sive, inconsistent in rigor, and variable in determining the qual-
ity of evidence. Morever, these processes are generally developed
by physicians for diagnostic minded physicians and not for the
analytically minded pharmacy practitioner. These practitioners
need a time-sensitive decision making process that allows them
to make firm decisions and recommendations based on results
of rigorously conducted clinical trials while incorporating their
own clinical judgment. Figur
Figuree 1.1 illustrates how the practitioner
must use caution in developing a recommendation and/or deci-
sion when this type of evidence does not exist.
Figur
iguree 1.1: Explanation of Differences in Firm and Cautious Recommenda-
tions Based on Trial Type
Practitioners need a time-sensitive decision
Key
making process that leads to evidence-supported
Idea decisions and recommendations.
Chapter 11: Introduction 5
The Solution
The University of Missouri–Kansas City School of Pharmacy Drug
Information Center developed the 5-Step Evidence-Based Medi-
cine Process that has been taught as a required course to Doctor
of Pharmacy (Pharm.D.) students for the last 10 years. This pro-
cess exhibits the following characteristics:
Offers less complexity
Offers time-sensitive decision making support by allevi-
ating time-intensive methods
Maintains rigor
Categorizes quality of the evidence in a simple, straight-
forward, and logical manner
Provides a process designed specifically for pharmacy
practitioners making drug therapy decisions
Figur
iguree 1.2: 5-Step Evidence-Based Medicine Process
This process involves five steps as illustrated in Figur
Figuree 1.2.
Step one is defining the clinical question. Defining the clinical
question is essential in providing direction to the remaining four
steps of this evidence-based medicine process. This step may be
the hardest one in the process because it involves the conversion
of a clinical problem into an answerable clinical question. A well-
constructed answerable clinical question clearly presents the true
clinical problem, provides guidance to pertinent evidence, and
suggests the format of the recommendation to solve the prob-
lem.
Step two involves searching the literature for articles associ-
ated with this clinical question. The assumption is that the reader
has completed a basic literature retrieval course. A comprehen-
6 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
sive discussion of resources for searching the literature is beyond
the scope of this book. Furthermore, most practitioners are too
busy to conduct extensive and sophisticated database searches to
ensure a comprehensive review of the pertinent literature. For
this reason, the practitioner must rely on specialists in drug in-
formation centers and health system libraries to conduct the
searches for them. Specific references will be identified for read-
ers who want a review of searching processes.
Step thr ee is evaluating literature found. Again, the assump-
three
tion is that the reader has completed at least a basic critical evalu-
ation course with some exposure to biostatistics. Although it is
not a requirement to understand the evidence-based medicine
process presented in this book, those readers without this back-
ground or those desiring a “refresher” review can refer to addi-
tional resources identified throughout the book.
Step four is determining the quality of identified and criti-
cally evaluated evidence.. Several different hierarchies of evidence
are available to accomplish this step.5–15 The categorization sys-
tem presented in this book provides a simple, straightforward,
and logical approach, which incorporates a modified technique
to determine the quality of evidence originally described in Drs.
Deborah Cook and Gordon Guyatt’s 1992 seminal work.1 This
fourth step serves as a bridge to the fifth step.
Step five is developing a conclusion and recommendation
with supporting justification. This step involves the creation of a
specific recommendation statement supported by the efficacy,
safety, and other special considerations/special populations pro-
vided by the evidence. Cost of therapy is also factored into this
final recommendation.
How This Book Will Help You
This book is not a text book, but rather a “how to” or self-devel-
opment type resource. The book teaches the practitioner how to
incorporate the 5-Step Evidence-Based Medicine Process into daily
drug therapy decision making.
Chapter 11: Introduction 7
Key This book is not a text book, but rather a
Idea "how to" or self-development type resource.
A consistent goal has been to develop an evidence-based
medicine process that is simple enough to integrate into
practitoners’ thought processes without giving up any rigor or
quality associated with good clinical decision making. Based on
results of studies to determine the effectiveness of this process,
this goal has been accomplished over the last 9 years of develop-
ment.16 Now, that same evidence-based medicine process is made
available to the practitioner through this book.
The format has been carefully considered, allowing the reader
to complete the book within a couple of evenings and immedi-
ately incorporate the process into current clinical practice. In
addition, the reader is encouraged to use this book as a resource
while applying the 5-Step Evidence-Based Medicine Process in
practice.
Key The format allows the reader to complete the
book in a couple of evenings and immediately
Idea
incorporate the process into current clinical practice.
Chapter 2: Basics for Interpretation is a high-level review of
basic tools and concepts associated with study design and se-
lected biostatistic principles. Emphasis is placed specifically on
the more pertinent knowledge in these two areas that is required
to understand and effectively perform the Evidence-Based Medi-
cine Process. For instance, only four biostatistic concepts are dis-
cussed:
1. Significance
2. Power
8 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
3. Types of data
4. Appropriate statistical tests for the type of data undergo-
ing analysis
For a more comprehensive review of biostatistics, the reader
should refer to the suggested references.
The actual 5-Step Evidence-Based Medicine Process is de-
scribed in Chapters 3 through 7. Each chapter addresses a spe-
cific step in the process. Examples are provided to further illus-
trate the concepts being taught. In addition, figures and tables
are included to reinforce visual learning of the concepts.
The actual 5-Step Evidence-Based Medicine Process
Key
is described in Chapters 3 through 7 with each
Idea chapter addressing a specific step in the process.
Chapters 8–10 identify special considerations and describe
clinical pharmacy practice applications for this process. Chapter
10 has been devoted to the differences in practicing evidence-
based medicine with dietary supplements compared to conven-
tional pharmaceuticals. The use of evidence-based medicine prac-
tice with dietary supplements is an area requiring greater atten-
tion. This chapter is an attempt to address that need.
At the end of this book, you will find a glossary of evidence-
based medicine terms to assist in learning the language associ-
ated with this discipline. In addition, there is a section dedicated
to forms and tables used by our students during the initial stages
of learning the 5-Step Evidence-Based Medicine Process. This
section is appropriately named Evidence-Based Medicine Tools
and will hopefully be a help.
Chapter 11: Introduction 9
References
1. Cook DJ, Guyatt GH, Laupacis A, et al. Rules of evidence
and clinical recommendations on the use of
antithrombotic agents. Chest. 1992; 102(4 suppl):305S–
311S.
2. Sackett DL, Rosenberg WC, Gray JAM, et al. Evidence-
based medicine: what it is and what it isn’t. BMJ. 1996;
312:71–72.
3. Portney LG, Watkins MP. Foundations of Clinical Research:
Applications to Practice. 2nd ed. Stamford, CT: Appleton
& Lange; 2000.
4. Eddy DM. Evidence-based medicine: a unified approach.
Health Affairs. 2005; 24(1):9–17.
5. Guyatt GH, Sackett DL, Sinclair JC, et al. Users’ guides to
the medical literature. IX. A method for grading health
care recommendations. Evidence-Based Medicine Work-
ing Group [published erratum appears in JAMA. 1996;
275(16):1232]. JAMA. 1995; 274:1800–1804.
6. McGovern DPB, Summerskill WSM, Valori RM, et al. Key
Topics in Evidence-Based Medicine. Oxford, UK: BIOS Sci-
entific Publishers Limited; 2001.
7. Heneghan C, Badenoch D. Evidence-based Medicine Toolkit.
Oxford, UK: Blackwell Publishing Limited; 2006.
8. Mayer D. Essential Evidence-Based Medicine. Cambridge,
UK: Cambridge University Press; 2004.
9. U.S. Preventive Services Task Force. Guide to Clinical Pre-
ventive Services. 2nd ed. Baltimore, MD: Williams &
Wilkins; 1996.
10. The Canadian Task Force on the Periodic Health Exami-
nation. The Canadian Guide to Clinical Preventive Health
Care. Ottawa: Health Canada; 1994.
11. Sackett DL. Rules of evidence and clinical recommenda-
tions. Can J Cardiol. 1993; 9:487–489.
10 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
12. Guyatt G, Rennie D, eds. User’s Guides to the Medical Lit-
erature. Chicago, IL: AMA Press; 2002.
13. Agency for Healthcare Research and Quality. Systems to
rate the strength of scientific evidence. Available at:
http://www.ahrq.gov/clinic/epcsums/strengthsum.pdf. Ac-
cessed April 6, 2007.
14. Scottish Intercollegiate Guidelines Network. SIGN 50: a
guideline developer’s handbook. Available at: http://
www.sign.ac.uk/guidelines/fulltext/50/section6.html. Ac-
cessed April 6, 2007.
15. National Institute for Health and Clinical Excellence.
Guideline development methods – Chapter 7: reviewing
and grading the evidence (revised March 14, 2005). Avail-
able at: http://www.nice.org.uk/page.aspx?o=247836.
Accessed April 6, 2007.
16. Pace HA, Norris KP, Bryant PJ. Evaluation of evidence-
based medicine education at the University of Missouri–
Kansas City School of Pharmacy. Unpublished data pre-
sented in poster format at the American Association of
Colleges of Pharmacy 2006 Annual Meeting in San Di-
ego, CA.
Chapter 2
Basics for
Interpretation
Patrick J. Bryant, Heather A. Pace,
Timothy A. Candy, & Antoine D.
Richardson
Why Should I Read This?
Essential Study Design Concepts
Interventional Trial Design
Observational Trial Design
Prospective
Retrospective
Case control
Results of observational trials
Key Concepts of Trial Design
Essential Biostatistic Concepts
Statistical Significance
Alpha value
P-value
Power
Appropriate Test for Data Analyzed
Types of data
Statistical tests applied to data
Summary
References
Why Should I Read This?
An understanding of some basic study design and statistical con-
cepts is necessary to effectively apply the 5-Step Evidence-Based
Medicine Process described in this book. This chapter is not in-
tended to be a comprehensive review of study design and biosta-
11
12 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
tistics. Rather than focusing on selecting and performing specific
study designs and statistical tests, this chapter describes practi-
cal applications of basic concepts in these two areas with an em-
phasis on interpretation of clinical research results. For those
practitioners who want a full review of either subject, the refer-
ences used for this chapter provide an excellent list of resources.1–
13
In addition, a glossary of terms frequently used in clinical re-
search articles is provided in the Glossary of Evidence-Based
Medicine Terms found in the back of this book.
Essential Study Design Concepts
Interventional Trial Design
An interventional study design, the most familiar study type, is
referred to as experimental study design. In this design, subjects
receive a treatment (or intervention) they would not otherwise
receive, and then the effect on a particular outcome is observed.
Subjects are randomly assigned to treatment groups by a ran-
domization scheme. If done correctly, subjects have an equal
chance of being assigned to a particular treatment group. All
interventional studies are prospective—that is, they look forward
in time. Interventional study designs are better at establishing
cause-and-effect relationships than observational designs because
the influence of confounding factors can be more controlled in
an interventional study.
Interventional studies can be either parallel or crossover de-
sign. In a parallel design, both groups are given treatment simul-
taneously and observed. Parallel studies are used with common
disease states where obtaining adequate numbers of participants
is not a problem. In addition, parallel group studies are used
when treatment effects are anticipated to continue after treatment
is discontinued, thus eliminating a crossover design as a possible
option (see Figur
Figuree 2.1
2.1). Crossover studies are often used to re-
duce the number of subjects needed to meet statistical power
because subjects serve as their own control. In a crossover de-
sign, subjects receive a particular treatment for a period of time
and then are switched to the alternate treatment. Crossover stud-
Chapter 22: Basics for Interpretation 13
Drug A
Run in
Drug B
Figur
iguree 2.1: Parallel Group Study Design
Drug A Drug A
Run in
Drug B Drug B
Figur
iguree 2.2: Crossover Study Design
ies must allow ample time for washout of the previous treatment
to ensure that treatment effects will not be carried over. The ef-
fects of each treatment are observed.
A crossover design can greatly reduce inter-subject variabil-
ity; therefore, the magnitude of effect can be more accurately cal-
culated in this type of study (see Figur
Figuree 2.2
2.2). While the cross-
over design has advantages, it is not appropriate in all disease
states. For instance, when mortality is the endpoint for a study, a
crossover design where patients are exposed to multiple treat-
ments would confound the results. In fact, patients may die be-
fore the crossover period and receive only one treatment. This
defeats the purpose of the design.
Another type of crossover trial known as a N of 1 trial in-
volves a single patient or small numbers of patients serving as
their own control who receive alternating courses of treatment
over a specific period of time. Because such a design involves
very small numbers of patients, these studies are limited by
generalizability but may be useful in rare disease states or situa-
tions in which funding is limited. Although these studies may
contain costs, this design often requires an increase in study pe-
riod to achieve the desired amount of treatment exposure in the
subjects. The study period is frequently doubled to allow the
14 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
subjects to serve as their own control. Caution should be used
when applying these results to the general population because
the number of patients exposed with this type of design is ex-
tremely limited.
Observational Trial Design (Epidemiological Design)
In observational trials, subjects are not assigned an intervention;
investigators merely observe subjects and gather information.
Subjects are chosen based on presence or absence of specific char-
acteristics. It is extremely difficult to control for confounding
variables in these studies. For this reason, cause and effect is dif-
ficult to prove and temporal relationships are not easy to estab-
lish; therefore, only associations can be observed. Results from
observational trials are less reliable than results from interventional
trials, but often times will lead to larger interventional studies to
examine effects closer in a better controlled environment. FigurFiguree
2.3 shows a diagram of the various observational study designs.
Prospective
In a prospective observational trial design, investigators define
the disease and the predicted variables prior to the onset of dis-
ease. Subjects are chosen based on specific characteristics, such
as smoking, and followed forward in time to observe the devel-
opment of a specific outcome such as lung cancer. If subjects are
followed forward in time, or prospectively, the groups are often
referred to as cohorts. One of the largest and well known cohort
studies is the Framingham Heart Study. These studies can also be
referred to as followup studies, meaning that they begin with a
cohort or group and follow the subjects forward in time until the
characteristic or disease is observed.
Retrospective
In retrospective observational trial design, investigators define
the disease and the predicted variables after disease onset. Retro-
spective cohort, also known as a trohoc study, examines existing
data and looks back in time to determine various characteristics
that may have led to disease development. For instance, investi-
gators examine the data for a group of subjects with a diagnosis
Chapter 22: Basics for Interpretation 15
Prospective Observational Trials
Smokers
6 Lung Cancer
6
Non-Smokers
Retrospective Observational Trials
Smokers 6
6 Lung Cancer
Non-Smokers
Case Control Studies
Lung Cancer 6 Smokers
6
6 Non-Smokers
6
No Lung Cancer 6 Smokers
6 Non-Smokers
Figur
iguree 2.3: Observational Study Designs
of lung cancer and analyze their smoking history to determine if
an association exists between smoking and development of lung
cancer.
Case Control
In a case control study, investigators look back in time and com-
pare two groups: the cases and controls. Cases are the group that
have a specific characteristic or disease, and the controls are cho-
sen as comparison and do not have the characteristic or disease.
The investigators aim to determine if there are any characteristics
that differ between the two groups such as a history of smoking.
In such a study, information can be gathered in various ways,
including survey, chart review, and direct interview. Case control
studies are useful to gather information regarding rare disorders
or disorders in which there is a long period between exposure
and development of outcome. Case control studies are especially
susceptible to bias, especially if investigators rely on the recall of
subjects.
16 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Results of Observational Trials
Results of observational trials are reported by measures of associa-
tion such as odds or risk ratios, all of which need to be interpreted
with caution and a good understanding of the definitions. Risk
ratios are calculated on data from prospective trials to determine
the risk of an outcome or adverse event in one group compared to
another group. Relative risk, relative risk reduction, and absolute
risk reduction are all types of risk ratios that need to be interpreted
with caution and a good understanding of the definitions.
Relative risk is the risk of an outcome in one group com-
pared to another (see Figur
Figuree 2.4
2.4). Relative risk reduction, on the
other hand, represents the relative change from baseline in out-
come or adverse event rate between intervention and control
group (see Figur
Figuree 2.5
2.5). Relative risk reduction may elevate the
benefit of a treatment because it does not account for the vari-
ability of baseline risk between subjects and assumes similar risks
for all. Absolute risk reduction is a better representation of the
magnitude of effect for a treatment because it takes into account
the baseline risks for individual subjects and represents the dif-
ference in adverse events between intervention and control groups
(see Figur
Figuree 2.6
2.6).
Outcome or Adverse Event (treatment group)
Relative Risk =
Outcome or Adverse Event (control group)
Figur
iguree 2.4: Relative Risk
Relative Risk Reduction = (1 – Relative Risk)
Figur
iguree 2.5: Relative Risk Reduction
Absolute Risk Reduction = (Risk in Control Group) –
(Risk in Treatment Group)
Figur
iguree 2.6: Absolute Risk Reduction
Chapter 22: Basics for Interpretation 17
Ultimately, what do all the numbers mean and how do the
results apply to patients? Number needed to treat provides a clear
illustration of the magnitude of benefit of a specific treatment by
providing the number of patients required to prevent one out-
come or adverse event (see Figur
Figuree 2.7
2.7).
1
Number Needed to Treat (NNT) =
Absolute Risk Reduction
Figur
iguree 2.7: Number Needed to Treat
Example 2.1 illustrates how different the numbers can
appear.
Example 2.1. Interpr eting Risk
Interpreting
A study was designed to compare simvastatin
(n=2221) versus placebo (n=2223) and incidence
of death from cardiovascular causes. The incidence
of death in the simvastatin group was n=182 and
placebo n=256.
• Relative Risk = death in simvastatin group ÷ death
in placebo group = 182 ÷ 256 = 0.71 (71%)
• Relative Risk Reduction = 1 – RR = 1 – 0.71 =
0.29 (29%)
• Absolute Risk Reduction = risk in placebo group
– risk in simvastatin group = 256/2223 – 182/
2221 = 0.033 (3.3%)
• Number Needed to Treat = 1 ÷ ARR = 1 ÷ 0.033
= 30
Key Concepts of Trial Design
Controls
• Placebo control – Placebo control trials demonstrate
if a treatment works, but do not establish superiority
18 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
over alternative treatments. Placebo controls can be
unethical, especially in patients with an established
disease such as diabetes. In such a case, an active con-
trol would be more appropriate.
• Active control – Active control trials demonstrate if a
treatment works and also aid in determining superi-
ority of certain treatments. Often times the “gold stan-
dard” therapy is used as the control.
• Historical controls – Historical controls are used when
new therapies have been proven better and it is deter-
mined that it would be unethical to use a previous
treatment as an active control. For instance, it has been
determined that an Alzheimer’s Disease drug has an
effect in reversing the disease. From this, the extent
of the effect needs to be determined, but current treat-
ments cannot be used because none of them actually
reverse the disease.
Blinding – Appropriate blinding techniques are impor-
tant to reduce bias; double-blind designs are ideal to en-
sure that neither the subject nor the investigator is af-
fected by the knowledge of the treatment received. Be-
cause it may be impossible to blind treatment in certain
situations, steps should be taken to ensure bias does not
affect the results of the trial. It is important to be aware of
the impact that blinding has and evaluate the results of
the trial for bias.
Randomization – Randomization attempts to ensure simi-
lar demographics and baseline characteristics and allows
confounding variables to be equal between treatment
groups.
Inclusion/exclusion criteria
• Appropriate inclusion criteria ensure subjects are in-
cluded for the target disease state and/or characteris-
tics for which the treatment is intended.
• Appropriate exclusion criteria are important to en-
sure the safety of subjects in the study.
Chapter 22: Basics for Interpretation 19
Outcomes – Appropriate outcome measurements are cru-
cial to applicability of the results of the trial; if outcome
measurements are not appropriate, it is difficult to apply
results of the trial to patient decisions.
Essential Biostatistic Concepts
Statistical Significance
In clinical trial design, a medical intervention and its effects are
usually compared to a different intervention, either a placebo or
an active comparator. In both cases, statistical differences could
exist between groups (e.g., demographic characteristics of the
study groups, therapeutic clinical outcomes, adverse event rates).
When these differences are statistically significant, the likelihood
of them existing due to chance is small.
Note that a calculated statistically significant difference is not
necessarily a clinically important difference between study groups.
For instance, a statistically significant difference may be shown
between study groups with diastolic blood pressures of 78 mmHg
and 81 mmHg; however, this would not necessarily be consid-
ered a clinically important difference.
Alpha value
The alpha (α) value is the threshold that, when crossed, indi-
cates a statistically significant difference between treatment groups
is reached. This level of significance is established by the study
investigators prior to the conduct of the study. The arbitrary α
value is commonly set at 5% (0.05). When the value is set higher,
such as 10% (0.10), it could be interpreted as a less rigorous
statistical analysis since the results would be presented in a more
favorable way. If the investigators set the value at 1% (0.01), the
statistical analysis would be interpreted as being more rigorous.
P-value
The p-value is an indicator to demonstrate statistically signifi-
cant differences between two treatment groups. The p-value rep-
resents the p robability that the statistical difference measured
20 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
between treatment groups occurred by chance alone and not the
intervention being studied. For instance, a p-value of 0.05 means
there is a 5% chance that a difference seen between treatment
groups occurred by chance alone. Explained another way, it means
there is a 95% chance that the measured difference observed be-
tween study groups is real and not due to chance. Statistical tests
can be used to determine a calculated p-value, and they are dis-
cussed later in this chapter.
Once alpha is set, a p-value that is determined from the sta-
tistical test applied to the data is generated and then compared to
the alpha value. If the p-value is less than the α level (e.g., p-
value < 0.05), the probability of the statistical difference being
attributed to random chance is very low (<5%). It indicates a
high likelihood that the intervention under study produced a
difference in the measure of interest (see Example 2.2 2.2).
Example 2.2. Determining Statistical
Significance
A study was designed to show that a new drug
was indeed more effective than the drug consid-
ered standard of care for a particular disease state.
The investigators set alpha at 0.05. This indicates
that the investigators are willing to accept a 5%
chance that if a statistically significant difference
is noted from this trial, that difference actually may
not really exist (Type I error). This is also referred
to as a false positive.
The correct statistical test was applied for the
type of data represented. The new drug showed
an improvement over the standard drug of 37%
for the primary outcome measurement. The sta-
tistical test generated a p-value of 0.0001
(p=0.0001). With the alpha set at 0.05, this p-
value is clearly less than the alpha level and, there-
fore, the difference noted is statistically significant.
Chapter 22: Basics for Interpretation 21
Additional information providing important points to remem-
ber regarding p-values and statistical significance can be found at
the end of this chapter.
Power
Power is the ability of a study to accurately detect actual differ-
ences between treatment groups, when such a difference truly
does exist. The higher the power is set for a study, the greater the
chance the study will show a difference between groups, if one
really exists. The lower the power is set, the greater the chance
the study will not show a difference between groups when one
actually exists.
It is also important to determine whether or not a per protocol
or intent-to-treat analysis was completed when determining whether
power is met. A per protocol analysis requires knowing the num-
ber of patients that completed the study. An intent-to-treat analy-
sis requires knowing the number of patients randomized into the
study groups. Note that a modified intent-to-treat analysis includes
specific criteria that must be met once a patient is randomized into
the study (e.g., must have received at least one dose of the study
drug or had at least one follow-up visit: see Example 2.3
2.3).
Additional information regarding the components that go into
calculating power can be found at the end of this chapter.
Example 2.3. P ower Is Met and No
Power
Differ ence Exists
Difference
A study was designed and conducted that showed
no statistically significant difference in the primary
outcome measurement between the two treatment
groups (p=0.70; alpha 0.05). Power was set at 80%
by the investigators during design of the trial. To
meet that power, 250 patients in each group were
required. When the study was completed, there
were 278 and 267 patients in each treatment
group, respectively. The investigators used a per
protocol analysis meaning they evaluated the out-
22 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
come measurement only in those patients com-
pleting the duration of the study as indicated in
the protocol. Based on this information, power
was met so there is an 80% chance that if a differ-
ence did exist, this trial would have shown that
difference.
If the investigators had set a power of 95% dur-
ing the design of the trial, this would have required
a sample size of 300 patients per group. Note that
if the investigators would have completed the study
with patient numbers of 300 or greater per group,
power would have been met. This situation would
have provided the investigators with a 95% chance
of the study showing a difference between groups,
if one truly existed.
The higher the power is set, the greater confi-
dence exists that the study will be able to correctly
detect a difference between groups, if one truly ex-
ists. In other words, a power of 95% that is met
gives the investigators a greater chance than the 80%
power to identify a difference, if one truly exists.
Sample size is the primary factor that an investigator can
directly control to achieve a study’s set power (see table in Fig-
ur
uree 2.8
2.8). Ironically, sample size is also the primary driver be-
hind the cost and time required to conduct a clinical study.
This can often contribute to the reason why many clinical tri-
als do not meet their required power. Enough time and/or
money could not be invested to assure a large enough sample
size to meet the set power.
The ideal situation is when the investigators set power, and
the sample size per group is adequately enrolled/completed to
meet that set power. In this case, whether there is a statistically
significant difference between treatment groups or not, there is
confidence that a difference would have been identified if one
actually existed (see Example 2.4
2.4).
Chapter 22: Basics for Interpretation 23
Sample Size
Needed to Power Met/
Situation Meet Power Not Met Conclusion
Power is set Identified in article Met 80% chance that a sta-
(>80%). and sample size tistically significant dif-
adequate to meet ference between groups
power. will be noted, if one
truly exists. Acceptable.
Power is set Identified in article Not Met <80% chance that a sta-
(>80%). but sample size not tistically significant dif-
adequate to meet ference between groups
power. will be noted, if one
truly exists. Not accept-
able.
Power is set Not identified in Not Met No easy way to verify if
(>80%). article. (assumption sample size was ad-
that must equate to meet power
be made) and detect a statistically
significant difference, if
one truly exists.
Power mentioned Identified in article, Not Met No easy way to verify if
but without details but unable to determine (assumption power was appropriate
(no mention of %). if sample size adequate that must (>80%).
to meet power since be made)
actual percentage not
revealed.
Power is not Not identified in Not Met No easy way to verify if
reported (no article and unable to (assumption sample size was
mention of %). determine if adequate that must adequate or power was
to meet power since be made) appropriate (>80%).
power not set.
Power is claimed Not identified in Not Met No easy way to verify if
to be met, but article and unable to (assumption sample size was
not reported (no determine if adequate that must adequate or power was
mention of %). to meet power since be made) appropriate (>80%).
no mention of actual %.
Figur
iguree 2.8: Various Ways that Power and Sample Size Can Be Reported
24 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Example 2.4. P ower Is Met and No
Power
Differ ence Exists
Difference
A study was designed and conducted that showed
no statistically significant difference in the pri-
mary outcome measurement between the two
treatment groups (p=0.85; alpha 0.05). Power
was set at 80% by the investigators during de-
sign of the trial. To meet that power, 250 pa-
tients in each group were required. There were
270 patients randomized into each treatment
group, respectively. The investigators used an
intent-to-treat analysis meaning they evaluated
the outcome measurement in all those patients
randomized into the study. Based on this infor-
mation, power was met and, therefore, we have
an 80% chance of observing a difference between
treatment groups, if one really did exist.
Note that if a difference would have been
shown in the above study, knowing if power was
met is not as crucial since enough patients com-
pleted the trial to actually show a difference.
If the results of a study conclude that no statistically signifi-
cant differences exist between treatment groups, then it is im-
perative to verify that power was set and met. The validity of the
study’s results becomes questionable if power was set but not
met with the necessary number of patients enrolled/completed
in the trial. In the following example (see Example 2.5
2.5), the study
did not have a large enough sample size to meet power and there-
fore, detect actual differences if they really existed. When this
happens, inaccurate conclusions can be made by the investiga-
tors that no difference exists when one really may exist.
Chapter 22: Basics for Interpretation 25
Example 2.5. P ower Not Met and No
Power
Differ ence
Difference Exists
A study was designed and conducted that showed
no statistically significant difference in the primary
outcome measurement between the two treatment
groups (p=0.16; alpha 0.05). Power was set at 80%
by the investigators during design of the trial. To
meet that power, 250 patients in each group were
required. When the study was completed, there
were 207 and 228 patients in each treatment
group, respectively. The investigators used a per
protocol analysis meaning they evaluated the out-
come measurement only in those patients com-
pleting the duration of the study as indicated in
the protocol. Based on this information, power
was not met and we cannot be sure that a differ-
ence may exist if enough patients would have been
entered into the study to meet power.
If the results concluded that a statistically significant differ-
ence did exist between treatment groups, power becomes less of
a concern because power is associated with an actual difference
being detected if one exists (see Example 2.6 2.6). The important
question in this situation is whether the study participants’ char-
acteristics are representative of the larger target population
planned for the treatment being tested. Clinical judgment is re-
quired to make the correct decision in this situation.
Example 2.6. P ower Not Met and
Power
Differ ence Exists
Difference
A study was designed and conducted that showed
a statistically significant difference in the primary
outcome measurement between the two treatment
groups (p=0.0012; alpha 0.05). Power was set at
80% by the investigators during design of the trial.
26 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
To meet that power, 250 patients in each group
were required. When the study was completed,
there were 233 and 241 patients in each treat-
ment group, respectively. The investigators used
a per protocol analysis meaning they evaluated the
outcome measurement only in those patients com-
pleting the duration of the study as indicated in
the protocol. Based on this information, power
was not met. However, enough patients were evi-
dently entered into the trial to show a statistically
significant difference so not meeting the set power
is less of a concern.
Figur
Figuree 2.9 illustrates various power scenarios that can oc-
cur when a statistically significant difference between groups does
or does not exist. In addition, the reliability of the study results
in each of these scenarios is provided.
Statistically significant difference between groups detected?
Yes No
Power not as great Power analysis performed?
of a concern
Yes No
Is sample size large
enough to represent the
general population? Power Met? Unable to assess if study
could detect a difference
if one existed (Major
Yes No Limitation)
Study powered to detect a Study not powered to detect a
difference (Major Strength) difference (Major Limitation)
Figur
iguree 2.9: Power Scenarios
Chapter 22: Basics for Interpretation 27
Appropriate Test for Data Analyzed
A study must use the appropriate statistical test for the type of
data being analyzed to assure the integrity of the results. To fully
understand this concept, a basic knowledge of data types and
data distribution is required.
Types of data
Data or variable scales consist of four different types of measure-
ment: interval, ratio, ordinal, and nominal. Figur
Figuree 2.10 provides
a definition summary table of each type of data and respective
data distribution.
Interval and ratio data are both considered “continuous” data
scales, having a known, equal distance between each data point.
Interval data scales can contain negative numbers. An example
includes temperature measured in the Celsius or Fahrenheit scale.
Ratio data scales contain no negative numbers. Examples of ratio
data include age, weight, serum glucose, blood pressure, and in-
cidence rates of outcomes. Ratio data can also be described in
“ratios” since there are no negative numbers. Even though tech-
nical differences exist, it is important to know that interval and
ratio data are handled the same way when selecting the most
appropriate statistical tests.
Ordinal data are data that can be ranked in a specific order.
This type of data differs from continuous data because the dis-
tance between data points are not equal. An example of ordinal
data is a Likert-type scale, where a patient’s mood is measured in
ranked order as no agitation, mild agitation, moderate agitation,
or extreme agitation.
Nominal data are also categorical data, but cannot be ranked
like ordinal data. Two subgroups of nominal data are binomial and
non-binomial. Nominal data that has only two possible outcomes
is called “binomial” data. For instance, binomial nominal data is
commonly used when measuring mortality and full recovery (ei-
ther it happened or it did not). Non-binomial nominal data has
more than two possible outcomes, but still cannot be ranked like
ordinal data. For instance, non-binomial nominal data includes
eye color (blue, brown, hazel) and various ethnicities.
28 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Type of
Type of Data Statistical
Data Definition Examples Distribution Test Used
Interval Continuous data Temperature Usually normal Parametric unless
scales with (Fahrenheit distribution the data is skewed;
known, equal and Celcius) (also known as then non-
distance between parametric parametric may be
each interval; distribution). appropriate.
they can contain
negative numbers.
Ratio Continuous data Age, weight, Usually normal Parametric unless
scales with known, serum glucose, distribution the data is skewed;
equal distance blood pressure, (also known as then non-
between each and incidence parametric parametric may be
interval; they rates of distribution). appropriate.
contain a non- outcomes
arbitrary, absolute
zero (i.e., no
negative numbers).
Ordinal Data that can Likert-type Non-normal Non-parametric
be ranked in pain scale distribution
specific order. where pain (also known as
The intervals is ranked as non-parametric
between data no pain, mod- distribution).
points are not erate pain, and
equal distance. extreme pain
Nominal Categorical data Eye color, Both nominal and Non-parametric for
that cannot be ethnicity, binomial nominal both nominal and
ranked. and gender data have binomial nominal.
non-normal
“Binomial” Mortality distribution
nominal data and full (also known as
has only two recovery non-parametric
outcomes. (either distribution).
happened
or did not)
Figur
iguree 2.10: Types of Data to Be Analyzed
Chapter 22: Basics for Interpretation 29
Each type of data represents a specific distribution when plot-
ted on a graph. See Figur
Figuree 2.11 for a visual example of each type
of distribution. The two most important types of distribution are
parametric (normal) distribution and non-parametric (non-
normal) distribution (see Example 2.72.7).
Example 2.7. Normal and Non-Normal
Data Distribution
An example of normal distribution would be to
plot the height of the students in a typical phar-
macy school class. If this sample of students had
evenly distributed characteristics, this plot could
represent the normal distribution of heights for
the entire college campus.
This same example can serve to illustrate a
non-normal distribution. This situation would be
to plot the height of the students in that same
pharmacy school class in addition to including
the basketball team. When looking at this distri-
bution of heights, one can see that the height is
not normally distributed. The addition of several
of the tallest students on the college campus has
skewed the distribution. The addition of a group
not representative of the overall college campus
confounds the normal distribution and distrib-
utes the height data in a non-normal manner.
Statistical tests applied to data
Statistical tests applied to data are described as being either para-
metric or non-parametric in nature. In general, continuous data
(interval and ratio) should be analyzed using parametric statisti-
cal tests, and categorical data (ordinal and nominal) should be
analyzed using non-parametric statistical tests. Each type of sta-
tistical test has an underlying set of assumptions to the data be-
ing analyzed. Parametric tests assume a normal distribution, while
non-parametric tests do not make assumptions about the distri-
30 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
20
10
0
0 10 20
Parametric (normal) Distribution
40
30
20
10
0
0 10 20 30
Positive Non-parametric (non-normal) Distribution
30
20
10
0
0 10 20 30
Negative Non-parametric (non-normal) Distribution
Figur
iguree 2.11: Parametric and Non-Parametric Data Distributions
bution of data. As long as these assumptions are met, then the
statistical tests can provide accurate results (see Figur
Figuree 2.12
2.12).
If an investigator uses a parametric statistical test for categorical
data, the results of that statistical test are questionable and one
must assess them cautiously in making any conclusive statements.
Technically, it is inappropriate to use parametric statistical tests
on categorical data. Parametric tests have an underlying assump-
tion of normal distribution using continuous data. Categorical
data have a non-normal distribution and, therefore, do not fit the
underlying assumption for the parametric test.
Chapter 22: Basics for Interpretation 31
In special situations, non-parametric tests may be used to
analyze continuous data such as when data has become non-
normally distributed or “skewed.” When this is noted, it should
be questioned why the investigators would use a non-parametric
test to analyze normally distributed continuous data.
In general, parametric statistical tests should be used for con-
tinuous data that is normally distributed (interval and ratio data).
Non-parametric statistical tests are used for types of data where
no assumption of normal distribution can or is being made.
The table in Figur
Figuree 2.13 assists the practitioner in determin-
ing which common statistical tests may be used for interval, ra-
tio, ordinal, and nominal data. If a statistical test is not listed in
the table, one should refer to other statistical resources listed as
references for this chapter. In the event that this strategy fails, a
quick search on the Internet using the statistical test in question
as the search term can also be useful.
The following Examples 2.8–2.10 provide an opportunity to
utilize the table found in Figure 2.13 and become familiar with the
above concepts of parametric and non-parametric data and tests.
Example 2.8. Appr opriate T
Appropriate est for
Test
Type of Data Analyzed
In a particular trial, patients were randomly as-
signed to one of two groups: Drug A or Drug B
group, respectively. To measure the effect that each
drug had on controlling blood glucose levels after
a meal, serum glucose levels were drawn at set
times after the meal. These serum glucose levels
were the primary outcome measure for this study.
The statistical methodology section stated that a
Student’s t-test was used to determine if a statisti-
cally significant difference was seen between Drug
A and Drug B in the reduction of serum glucose
levels after the 3-month trial.
In this example, serum glucose levels are the
data being collected as the primary outcome.
32 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Type of Data: (serum glucose levels) Continuous
data having a known, equal distance between each
interval and containing a non-arbitrary, absolute
zero (i.e., no negative numbers). This is consid-
ered ratio data
data.
Appr opriate T
Appropriate est: (Student’s t-test) Yes, accord-
Test:
ing to the table in Figure 2.13; Student’s t-test (a
parametric test) can be used with ratio data (nor-
mal distribution).
What type of data is being analyzed?
Continuous Categorical
Ratio or interval level data Ordinal or nominal level data
Is the data assumed to be
normally distributed?
Yes No
Was the data analyzed
using parametric Was the data analyzed using
statistical tests? non-parametric statistical tests?
Yes No
Did the results detect
a statistically Yes No
significant difference?
Yes No
This study’s data The validity of the data analysis
analysis is likely valid. must be questioned as the
statistical test is not appropriate
for the data being analyzed.
Figur
iguree 2.12: Statistical Test Decision Flowchart
Chapter 22: Basics for Interpretation 33
Continuous
Nominal Ordinal
Ordinal (Ratio or Inter val)
Interval)
• Chi-Square test (x2) • ANCOVA ranks • ANCOVA
• Cochran’s Q test • ANOVA ranks • ANOVA
• Fischer’s exact test • Friedman ANOVA • Pearson
• Logistic Regression • Kendal Rank correlation Correlation
coefficient Coefficient (r)
• Mantel-Haenszel
• Kruskal-Wallis ANOVA • Student’s t-test
• McNemar’s test
• Mann-Whitney U test
• Spearman Rank correla-
tion coefficient
• Wilcoxin Rank Sum test
• Wilcoxin Signed Rank
test
Figur
iguree 2.13: Commonly Used Parametric and Non-Parametric Statistical
Tests by Type of Data Analyzed * Note that each of these tests has its own
underlying assumptions and are sometimes meant for very specific situations,
depending upon the study design and type of data being analyzed. Please refer to
statistical textbook references to learn more about these specific tests and when
they are appropriately applied.
Example 2.9. Inappr opriate T
Inappropriate est for
Test
Type of Data Analyzed
A study was conducted to measure an investiga-
tional drug’s effect on decreasing agitation in bi-
polar children. The comparison group was stan-
dard therapy. Each child’s caregiver was asked to
rate the amount of agitation at set times over the
8-week trial as no agitation, mild agitation, mod-
erate agitation, or severe agitation. The statistical
test used to determine if there was a statistically
significant difference between the two drugs for
reducing agitation was a Chi-square test.
In this example, the primary outcome mea-
sure was a Likert scale measuring the degree of
agitation.
34 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Type of Data: (Likert scale) Categorical data that
can be ranked in a specific order and the intervals
between data points are not equal in distance. This
is considered ordinal data.
Appr opriate T
Appropriate est: (Chi-square test) No, accord-
Test:
ing to the table in Figure 2.13; Chi-square test
should be used with nominal data, not ordinal
data. Note that a non-parametric test was being
used for data that is non-normal in distribution;
however, additional assumptions made with this
type of test (used with categorical data that can-
not be ranked) did not fit the data being analyzed
which was categorical and ranked data.
Example 2.10. Inappr opriate T
Inappropriate est for
Test
Type of Data Analyzed
In a trial designed to show if the standard of
therapy prevented the incidence of myocardial
infarctions, hospitalizations, and/or death, patients
on this standard of therapy were followed over a
3-year period after angioplasty and stent place-
ment. Patient or patient’s family member com-
pleted a questionnaire provided monthly asking
if the patient experienced a myocardial infarction
or hospitalization due to a cardiovascular event.
In addition, caregivers were asked in this ques-
tionnaire if the patient was still alive or not. The
statistical section stated that a two-way analysis
of variance (ANOVA) was used to analyze these
primary outcome measures.
The primary outcome measures were whether
a patient experienced a myocardial infarction, hos-
pitalization, and/or death.
Chapter 22: Basics for Interpretation 35
Type of Data: (occurred or didn’t occur) Categori-
cal data that cannot be ranked and has only two
outcomes (either it happened or it did not). This
is binomial nominal data.
Appr opriate T
Appropriate est: (Two-way ANOVA) No, accord-
Test:
ing to the table in Figure 2.13; this test (paramet-
ric test) is not used with nominal (non-paramet-
ric) data.
In general, parametric statistical tests should
be used for continuous data (interval and
Key ratio data) that is normally distributed.
Idea Non-parametric statistical tests can be used
for other types of data where no assumption
of normal distribution can or is being made.
Summary
This chapter describes practical applications of basic concepts
required for interpretation of clinical research results. Various
study designs are used in clinical research. Understanding the
type of study design described in an article allows identification
of strengths and weaknesses within the design that could affect
accurate interpretation of the results.
Correct interpretation of biostatistics applied to the results of
a study can sometimes be difficult. Knowing the relationship be-
tween alpha and p-values allows confirmation of claims being
made by the investigators. In addition, knowing that a study is
adequately powered provides confidence in results showing no
difference between treatment groups. Finally, being able to deter-
mine if the appropriate statistical test is used for the type of data
being analyzed gives assurance that reported statistically signifi-
cant differences really do exist between treatment groups.
Understanding these concepts is required to work the 5-Step
36 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Evidence-Based Medicine Process described in this book. With-
out a working knowledge of these essential concepts in study
design and biostatistics, practitioners could inaccurately inter-
pret trial results that may lead to inappropriate and potentially
unsafe clinical decisions.
References
1. Norman GR, Streiner DL. PDQ Statistics. 3rd ed. Hamilton,
London (Ontario): BC Decker Inc.; 2003.
2. Elenbass RM, Elenbass JK, Cuddy PG. Evaluating the
medical literature. Part 2: Statistical analysis. Ann Emerg
Med. 1983; 12(10):610–620.
3. Gaddis ML, Gaddis GM. Introduction to biostatistics: Part
1, basic concepts. Ann Emerg Med. 1990; 19:86–89.
4. Gaddis GM, Gaddis ML. Introduction to biostatistics: Part
2, descriptive statistics. Ann Emerg Med. 1990; 19:309–
315.
5. Gaddis GM, Gaddis ML. Introduction to biostatistics: Part
3, sensitivity, specificity, predictive value and hypothesis
testing. Ann Emerg Med. 1990; 19:591–597.
6. Gaddis GM, Gaddis ML. Introduction to biostatistics: Part
4, statistical inference techniques in hypothesis testing.
Ann Emerg Med. 1990; 19:820–825.
7. Gaddis GM, Gaddis ML. Introduction to biostatistics: Part
5, statistical inference techniques for hypothesis testing
with non parametric data. Ann Emerg Med. 1990; 19:1054–
1059.
8. Gaddis ML, Gaddis GM. Introduction to biostatistics: Part
6, correlation and regression. Ann Emerg Med. 1990;
19:1462–1468.
9. Riegelman RK. Studying a Study and Testing a Test. 4th ed.
Philadelphia, PA: Lippincott Williams & Wilkins; 2000.
10. Gehlbach SH. Interpreting the Medical Literature. 4th ed.
New York, NY: McGraw-Hill; 2002.
Chapter 22: Basics for Interpretation 37
11. Friedman LM, Furberg CD, DeMets DL. Fundamentals of
Clinical Trial. 3rd ed. New York, NY: Springer; 1998.
12. Glantz SA. Primer of Biostatistics. 6th ed. New York, NY:
McGraw-Hill; 2005.
13. Hulley SB, Cummings SR, Browner WS, et al. Designing
Clinical Research. 3rd ed. Philadelphia, PA: Lippincott
Williams and Wilkins; 2007.
38 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Significance and p-Values
The following are important points to remember regarding p-
values and statistical significance:
1. P-values cannot be used as absolute proof that an inter-
vention has a particular benefit or not. The only way to
fully eliminate random chance as a contributor to study
results is to enroll the entire worldly population being
studied. Also, unknown confounding variables that have
nothing to do with the intervention(s) being studied could
have contributed to the measured statistical differences.
This is why proper study design to minimize confound-
ing variables is so important.
2. No matter how small the difference, any difference of ef-
fect between groups can be considered statistically sig-
nificant if the study design’s power is high enough (see
section on Power in this chapter).
3. When attempting to compare various treatments and their
associated effects based on clinical trial results, it is not
appropriate to compare p-values as a way to determine
which intervention is safer or more efficacious. P-values
do not equate to levels of magnitude.
Chapter 22: Basics for Interpretation 39
Components of Power
Power is related to another concept, the Beta (β) Level. The β
level is the probability of making a Type II Error, or falsely deter-
mining that no actual difference exists between two study groups
when a difference truly does exist (i.e., “false negative”). For ex-
ample, a β level of 20% (or 0.2) means that there is a 20% prob-
ability that the study will falsely determine that no difference
exists between groups when a difference truly does exist. How is
β related to power? Conceptually, power can be calculated as 1 –
β. Using our previous example of β set at 20%, then the resultant
power of the study is calculated as 1 – 0.2, which equals 0.8 or
80%. A study having an estimated power of 80% means that the
study has an 80% chance of seeing a difference between treat-
ment groups, if a difference really exists. Power set at 80% is
consider
considered ed appr opriate for most studies
appropriate studies; however, often 85%
to 90% power is set for high investment studies (e.g., long-term,
large trials, etc.).
While 1 – β is a very simplistic way of calculating power,
complex equations are used when attempting to quantify the true
statistical power of a study based on several factors. Components
that go into a power calculation include:
Level of significance (α) – usually set at 0.05
Type II Error Rate (β) – usually set at 0.20
Magnitude of difference in an outcome measure between
treatment groups (δ) – usually obtained from previous stud-
ies using the intervention if it exists; if not, then much more
speculative
Variability (σ)
Sample size (n)
These are the primary components of a power calculation. There
are several other components that may be considered by experi-
enced biostatisticians that involve far more complex concepts than
what is covered within the scope of this book.
Chapter 3
Define the
Clinical Question
Patrick J. Bryant
Step 1 – Define the Clinical Question
Framing the Clinical Question Using PICO
Effects on Other Evidence-Based Medicine Activities
Tips for Defining the Clinical Question
Problems to Be Avoided
Summary
References
Step 1 – Define the Clinical Question
The first step of the 5-Step Evidence-Based Medicine Process,
Define the Clinical Question, may be the most difficult one be-
cause it involves the translation of a clinical problem into an an-
swerable clinical question. Defining and structuring an appro-
priate clinical question are both critical. According to a survey
conducted by the Drug Information Service at the Medical Col-
lege of Virginia Hospitals, the original clinical question asked was
significantly different from the actual question that needed to be
answered 85% of the time.1 This problem suggests the need for
applying some type of systematic process to ensure the most ac-
curate definition of the clinical question. One approach using a
logical and systematic manner to define the problem and clarify
the clinical question has been proposed by drug information spe-
cialists.2 The goal of this approach is to understand the context
and scope of the problem.
41
42 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
85% of the time the original
Key clinical question identified is significantly
Idea different from the actual question
needing to be answered.
Framing the Clinical Question Using PICO
Accurately defining the clinical question is essential to providing
direction for the remaining four steps of the evidence-based medi-
cine process outlined in this book. Those steps include retriev-
ing the pertinent literature, critically evaluating the literature, cat-
egorizing the quality of the evidence, and developing a recom-
mendation that can be justified. A generic, minimally defined
clinical question does not provide the required direction for these
steps.
Example 3.1. Using Minimal Detail to
Define the Clinical Questions
An example of a generic, minimally detailed
clinical question is taking a newly marketed non-
injectable insulin product and asking if this
product should be added to a preferred drug list
within an institution. The following can be
determined from this minimal information:
1. context – a preferred drug list formulary
management activity for an institution
2. scope – a population-based decision for a
specific institution that may have a particular
patient population such as indigent and/or
Medicaid
With this example, only the context and scope of
the question are provided. The details necessary
to define the true clinical question are missing.
Using the PICO approach, the definitive details of
the clinical question can be determined.
Chapter 33: Define the Clinical Question 43
Accurately defining the clinical question provides
Key direction to the remaining four steps of the
Idea evidence-based medicine process.
An accepted approach for framing the clinical question is pro-
vided by Sackett and colleagues using an acronym approach re-
ferred to as PICO.3–5 This approach includes the following com-
ponents:
P atients – What individual patient or patient population is
associated with the clinical question? Are there any pa-
tient subgroups that require special attention?
I nterventions – What interventions can be used in this clini-
cal situation?
Comparison – What additional interventions can be consid-
ered and compared to the chosen intervention?
Outcome – What is the end result most desired for the indi-
vidual patient, patient population, and/or patient subgroup?
The specific patient group, therapeutic options to consider,
and desired results of using a specific therapeutic option must be
identified when defining the clinical question. Figur
Figuree 3.1 (see
also the “EBM Tool Kit” at the end of this book) details the PICO
process and the resulting benefits of using this approach.
An accepted approach for framing the clinical question
resulting in a well-defined clinical question is using
Key an acronym approach referred to as PICO. This
Idea acronym stands for Patient, Intervention,
Comparison, and Outcomes.
44 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
PICO
Patients
Initial
Interventions
Question
(no detail
state)
Comparison
- Context
- Scope
Outcomes
Figur
iguree 3.1
3.1: PICO Illustration
Example 3.2. PICO Appr oach Defined
Approach
The Patient population includes both type 1 and
2 diabetics either just initiating therapy or having
been on insulin for a period of time. A special
subgroup of patients to consider might include
those with a “needle phobia” or those who want a
more practical way to administer insulin when out
in public.
The I ntervention is the non-injectable insulin.
Related questions would further define how the
intervention is administered and provide a
description of the device needed for admin-
istration, frequency of administration, and dosing
equivalency to injectable insulin.
Chapter 33: Define the Clinical Question 45
The Comparison includes injectable insulins
and other interventions such as competitor non-
injectable insulins, insulin stimulators, and insulin
enhancers. Also a review of alternatives to the non-
injectable insulin represented by the I ntervention
is performed.
The Outcome is adequate glycemic control as
measured by fasting blood glucose levels and
HbA1C without the problems and inconveniences
Figuree 3.2
associated with injectable insulin (See Figur 3.2).
Using this PICO approach now allows development of a well-
defined clinical question as illustrated in Example 3.3
3.3.
Example 3.3. The W ell-Defined
Well-Defined
Clinical Question
“Are there safety and/or convenience benefits
associated with this non-injectable insulin
compared to injectable insulin, other non-
injectable insulins, insulin stimulators, or insulin
enhancers to provide normal blood glucose and
glycemic control in type 1 and 2 diabetic patients?
Is there a difference in patient motivation to use
the non-injectable insulin between patients just
initiating versus maintaining insulin therapy and/
or those patients with ‘needle phobia’?”
Effects on Other Evidence-Based Medicine Activities
Once the clinical question has been accurately defined, the fol-
lowing activities associated with the evidence-based medicine
process can be initiated.
46
PICO Benefits
Patients: - type 1 & 2 diabetes
The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
- initiating insulin Identify
therapy Search Terms
- maintaining insulin
therapy
- subgroup = “needle Determine
phobia” Relevance of
Defined Search Results
Initial Intervention: - non-injectable insulin
Clinical
Question - inhaled insulin
Focus Information
(no detail - large device-inconvenience Question Retrieval
state) - same dosing frequency (detailed
- requires dosing state)
conversion (IU to mg) Focus Critical
Comparison: - injectable insulins Evaluation
- non-injectable insulins
- Context - insulin stimulators Develop
- Scope Recommendation
- insulin enhancers with Justifications
- other alternatives Efficiently and
Outcomes: - normal range fasting Effectively
blood glucose
- normal range HbA1C
Figur
iguree 3.2
3.2: PICO Applied Illustration
Chapter 33: Define the Clinical Question 47
Identify specific search terms for use in retrieving perti-
nent information (see Example 3.4)
3.4).
Example 3.4. Specific Sear ch T
Search erms
Terms
• The specific intervention’s trade name
• Names of other competing non-injectable
insulins
• Names of insulin stimulators
• Names of insulin enhancers
• Names of injectable insulins
• Type 1 diabetes
• Type 2 diabetes
• Fasting blood glucose
• HbA1C
• “Needle phobia”
Determine the relevance of the search results now that
the details of the patient population, intervention, com-
parisons, and desired outcomes are known.
Focus information retrieval and critical evaluation efforts
toward the pertinent evidence as the specific patient popu-
lation, intervention, comparison, and desired outcomes
have been identified by the defined clinical question.
Develop a recommendation more efficiently and effectively
based on observations made possible by a well-defined
clinical question during the critical evaluation of the evi-
dence (see Example 3.5)
3.5).
48 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Example 3.5. Specific Obser vations
Observations
• Determine if there are safety and/or
convenience benefits associated with using
non-injectable insulin versus identified
alternative therapies.
• Determine if there is a difference in patient
motivation to use a non-injectable insulin
between type 1 and type 2 diabetic patients.
• Determine if there is a difference in patient
motivation to use a non-injectable insulin in
patients initiating injectable insulin therapy for
the first time versus patients already
maintained on injectable insulin.
• Consider if a subgroup of patients with “needle
phobia” is more likely to utilize the non-
injectable insulin compared to those without
this problem.
A well-defined clinical question assists in identifying
Key search terms, determining relevance of the search
Idea results, performing a critical evaluation of the
evidence, and developing a recommendation.
Tips for Defining the Clinical Question
Important points to consider when defining the clinical question
include the following:
Describe a group of patients with similar characteristics
as those in the problem or situation you are trying to de-
fine with the clinical question.
Chapter 33: Define the Clinical Question 49
Identify the primary intervention.
Identify the most likely alternative intervention(s) to be
considered.
Identify the primary desired outcome and how that out-
come is measured in the clinical setting.
Problems to Be Avoided
Finally, some common problems are associated with defining the
clinical question that should be avoided:
Complex clinical situations resulting in numerous ques-
tions identified can be handled by dissecting the situa-
tion into smaller, less complex, and more manageable
problems requiring fewer questions.
Too little or complete lack of adequate background infor-
mation can be handled by further questioning key people
associated with the clinical situation.
Lack of time to complete an evidence-based medicine
analysis can be due to an overabundance of clinical prob-
lems associated with the situation. Judicious prioritization
identifying those clinical problems having the greatest
impact on a patient or institution is one way to deal with
this issue.
Summary
Defining the clinical question must be done accurately. In addi-
tion, this step must be completed with enough detail to provide
the required information necessary to conduct the other four steps
of the evidence-based medicine process described in this book.
An acronym-based approach, referred to as PICO, can be used to
assist in defining the clinical question. The PICO approach pro-
vides a well-defined clinical question. This clinical question as-
sists in identifying search terms for retrieval of pertinent infor-
mation and in determining relevance of search results. In addi-
tion, focusing information retrieval, focusing critical evaluation
efforts, and maximizing efficiency and effectiveness of recommen-
50 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
dation development is accomplished with a well-defined clinical
question. Clearly, defining the clinical question accurately is the
foundation to the entire evidence-based medicine process.
References
1. Kirkwood CF, Kier KL. Modified Systematic Approach to
Answering Questions. In: Malone PM, Kier KL, Stanovich
JE, eds. Drug Information: A Guide for Pharmacists. 3rd ed.
New York, NY: McGraw-Hill Companies, Inc; 2006.
2. Calis KA, Sheehan AH. Formulating Effective Responses
and Recommendations: A Structured Approach. In:
Malone PM, Kier KL, Stanovich JE, eds. Drug Information:
A Guide for Pharmacists. 3rd ed. New York, NY: McGraw-
Hill Companies, Inc; 2006.
3. Sackett DL, Richardson WS, Rosenberg WMC, et al. Evi-
dence-based Medicine: How to Practice and Teach EBM. 2nd
ed. London: Churchill-Livingstone; 2000.
4. Richardson WS, Wilson MC, Nishikawa J, et al. The well-
built clinical question: a key to evidence-based decisions.
ACP J Club. 1995; 123:A12–A13.
5. Oxman AD, Sackett DL, Guyatt GH. Users’ guide to the
medical literature. JAMA. 1993; 270:2093–2097.
Chapter 4
Retrieve Pertinent
Information
Heather A. Pace, Lindsey N. Schnabel,
Morgan L. Sperry, & Patrick J. Bryant
Step 2 – Retrieve Pertinent Information
Deciding Where to Start
Creating the Search Strategy
Broad to narrow search
Bibliographic searching
Organizing Search Results—Search and Sift Technique
Using the Internet as a Resource
Summary
References
Step 2 – Retrieve Pertinent Information
The second step of this 5-Step Evidence-Based Medicine Process,
Retrieving Pertinent Information, involves creating a comprehen-
sive search strategy to identify all of the pertinent evidence avail-
able relative to the clinical question. This information can be re-
trieved from a variety of databases and compendia. To complete
this step, the pharmacy practitioner needs a basic understanding
of common databases and standard search strategies. An incom-
plete search strategy may cause the omission of key information
and may result in an inappropriate recommendation affecting the
treatment and/or safety of the patient. Once this strategy is defined
and completed, the next step is to refine the search results. Search
strategies and the techniques used to organize search results will
be discussed in this chapter. Retrieving Pertinent Information is
an extremely important step in the 5-Step Evidence-Based Medi-
cine Process because it builds the foundation for this process.
51
52 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
An incomplete search strategy can result in an
Key
inappropriate recommendation directly
Idea affecting the patient’s care.
Deciding Where to Start
To ensure that recommendations are supported by the best avail-
able evidence, it is important for a clinical practitioner to under-
stand the steps used to provide a successful search. An effective
search strategy begins broad with the use of tertiary resources
and narrows to primary resources. Tertiary resources include text-
books and compendia (e.g., Micromedex, LexiComp, Clinical
Pharmacology, Facts and Comparisons). These resources are used
for gathering general background information pertaining to the
clinical question.
Secondary resources are indexing and abstracting services that
simplify the search and identify primary literature. Some examples
include PubMed (or Medline), International Pharmaceutical Ab-
stracts, Iowa Drug Information Services, and EMBASE. Primary
resources refer to journals containing original research, includ-
ing randomized controlled trials, review articles, case studies, and
meta analyses. The resources discussed above are crucial in car-
rying out the 5-Step Evidence-Based Medicine Process.
Many references mentioned in this chapter are not available
to the clinical practitioner; however, they are often available in
drug information centers and health science libraries. For this
reason, it is important to develop a relationship with a drug in-
formation specialist or medical librarian to ensure a comprehen-
sive and high quality search.
For a general review of resources and searching techniques,
the reader is directed to the references in this chapter. These ref-
erences provide a comprehensive review of medical information
sources such as commercially available databases and directions
for searching them.
Chapter 44: Retrieve Pertinent Information 53
Creating the Search Strategy
Broad to narrow search
A precise search strategy is critical to ensure all pertinent evi-
dence is identified.17,18 Too much or too little detail in the search
strategy can result in limited usable evidence. For instance, spe-
cific search terms can limit the results while the use of more gen-
eral search terms often provides excessive amounts of non-rel-
evant information.
Example 4.1. Using General and
Specific Sear ch T
Search erms
Terms
The defined clinical question is whether Drug A
is as effective as Drug B regarding improvement
of bone density in the treatment of osteoporosis.
Specific Search T
Search er
Terms:
erms:
Drug A brand name
Drug B brand name
General Search T
Search er
Terms:
erms:
Drug A generic name
Drug B generic name
Osteoporosis
Bone Density
Drug A—all approved indications besides
osteoporosis
Drug B—all approved indications besides
osteoporosis
1. Use of specific search terms listed above can
limit the search to those articles where only
the brand name appears. More possible “hits”
54 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
may appear if the search is made somewhat
more general by searching both the brand and
generic names of Drug A and Drug B.
2. Searching by brand and generic names of Drug
B could result in too general of a search if Drug
B has been on the market long enough to lose
patent exclusivity. In this case, not all the ge-
neric products identified with this search are
going to be pertinent to the situation. For in-
stance, if the brand name product is the only
oral form and this is the route pertinent to the
clinical question, then only the brand name
drug would be of interest.This is often the case
with standard or accepted treatment.
Bibliographic searching
Once the original search is complete, a technique referred to as
bibliographic searching can aid in optimizing results of the search.
The technique involves reviewing citations of previously identi-
fied pertinent information in the original search. This technique
can help identify additional literature as well as help in discover-
ing additional search terms that may be useful to broaden the
original search. Bibliographic searching helps create a network
or “spider web” of information relating to the subject and helps
ensure search results are thorough and complete, thus leading to
a strong and confident recommendation.
Key Bibliographic searching ensures
Idea thorough and complete search results.
Chapter 44: Retrieve Pertinent Information 55
Organizing Search Results—Search and Sift Technique
Applying the skills in Step 4 – Categorize the Quality of Evi-
dence to the retrieval process can maximize efficiency of the
literature search and make an overwhelming list of search re-
sults manageable. A technique referred to as “Search and Sift”
helps the clinician prioritize and organize search results by
the quality of evidence and allows evidence to be eliminated
that is not pertinent to the clinical question.
The technique involves “sifting” through the abstracts of search
results using key words identified from the Ten Major Consider-
ations to quickly determine the quality of evidence (see Chapter
5: Step 3 – Evaluate Literature). This technique also helps elimi-
nate lower quality, less reliable evidence. It is another way in which
the 5-Step Evidence-Based Medicine Process outlined in this book
helps optimize the clinical decision making and recommenda-
tion process (see Figur
Figuree 4.1
4.1).
Initial Sift Results
(Apply Steps 3 and 4 to each
article from search results)
Starting Point Final Sift
Randomized Results
Controlled Trials
SEARCHING Review Articles Randomized Best
Controlled Trials quality of
Meta Analysis evidence
Online Literature
Sift Meta Analysis Sift pertinent
Searching Case Series/Case to the
Reports Case Series defined
clinical
Abstracts Abstracts question
Consensus
Statements
Figur
iguree 4.1. “Search and Sift” Strategy
56 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Example 4.2. Using the “Sear ch and
“Search
Sift” Technique
Sift” Technique
As a practitioner, you have been asked to provide
guidance on the prophylaxis of stress ulcers. Af-
ter identifying and reviewing the available thera-
peutic guidelines on stress ulcers, you determine
the need to update the original evidence-based
guidelines published in 1998. You will need to
first retrieve the pertinent information that has
become available since this guideline was pub-
lished.
To help you with this project, you contact a
colleague in the drug information center and re-
quest a search of the literature for anything that
could be used to update this existing 1998 guide-
line. You receive results of an online literature
search. At this point, you also perform a biblio-
graphic search. Because you are proficient using
your evidence-based medicine skills, you employ
the “Search and Sift” technique and are able to
review the abstracts to determine the quality of
evidence of the studies and then eliminate evi-
dence that will not help form and support a
strong recommendation. You are able to quickly
identify six pertinent articles from the 25 results
that your colleague identified with the original
search (see Figur
Figuree 4.2
4.2).
A technique referred to “Search and Sift” helps the
Key clinician prioritize and organize search results by
Idea quality of evidence and allows evidence to be
eliminated that is not pertinent to the clinical question.
Chapter 44: Retrieve Pertinent Information 57
Figur
iguree 4.2. “Search and Sift” Strategy for Stress Ulcer Practice Guideline
Update
Using the Internet as a Resource
Search results available from various Internet search engines, such
as Google, should be used with caution. Because the Internet is
so widely available, the source of information is not always clear
nor is the information always accurate and/or reliable. A good
example is Wikipedia, an online “dictionary” which allows any
individual to contribute regardless of the source or validity of
information. Additionally, not all medical literature is indexed
on the Internet, further limiting the results of the search. In re-
mote cases, a general Internet search may be useful; however,
caution is advised. Results from the Internet should be reserved
as the last resort only after an exhaustive search of appropriate
databases has been completed.
Several factors should be considered before using informa-
tion obtained from the Internet in an evidence-based medicine
analysis16:
1. Documented medical evidence is present with citations,
not just opinions
58 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
2. Website credibility
3. Author’s credibility
4. Date of publication
5. Unbiased tone used
6. Peer reviewed
Key The Internet should be reserved as the
last resort only after an exhaustive search of
Idea
appropriate databases has been completed.
Summary
Retrieving the most pertinent information to be used in the 5-
Step Evidence-Based Medicine Process is crucial to making the
most accurate recommendation. This is important because these
recommendations affect the health and safety of patients. Today’s
practitioner must rely on drug information specialists and medi-
cal librarians to assist with this step. This will ensure the most
accurate and comprehensive search results without consuming
extensive time from the practitioner. Additional articles found in
the reference citations of studies identified by the search can in-
crease the practitioner’s confidence that a comprehensive search
has been achieved. To optimize efficiency of the search, the prac-
titioner should adopt the “Search and Sift” technique to priori-
tize and organize search results by quality of evidence. Finally,
caution must be used when including information obtained di-
rectly from the Internet.
References
1. Shields KM, Lust E. Drug Information Resources. In:
Malone PM, Kier KL, Stanovich JE, eds. Drug Information:
Chapter 44: Retrieve Pertinent Information 59
A Guide for Pharmacists. 3rd ed. New York, NY: McGraw-
Hill Companies, Inc; 2006.
2. Malone PM. Electronic Information Management. In:
Malone PM, Kier KL, Stanovich JE, eds. Drug Information:
A Guide for Pharmacists. 3rd ed. New York, NY: McGraw-
Hill Companies, Inc; 2006.
3. Greenhalgh T. How to Read a Paper. Oxford, UK: Blackwell
Publishing Ltd; 2006.
4. Brice A, Palmer J, Bexon N. Information Sourcing. In:
Hamer S, Collinson G, eds. Achieving Evidence-based Prac-
tice: A Handbook for Practitioners. 2nd ed. London, UK:
Elsevier Limited; 2005.
5. Chiquette E. Searching the Biomedical Literature: Find-
ing a Needle in a Haystack. In: Chiquette E, Posey LM,
eds. Evidence-Based Pharmacotherapy. Washington, DC:
American Pharmacists Association; 2007.
6. McManus RJ. Sources of Information. In: McGovern DPB,
Valori RM, Summerskill WSM, et al., eds. Key Topics in
Evidence-based Medicine. Oxford, UK: BIOS Scientific Pub-
lishers Limited; 2001.
7. Safranek S, Dodson S. Strategies for Finding Evidence.
In: Geyman JP, Deyo RA, Ramsey SD, eds. Evidence-based
Clinical Practice: Concepts and Approaches. Boston, MA:
Butterworth Heinemann; 2000.
8. Pirozzo S. Searching the Medical Literature. In: Mayer D,
ed. Essential Evidence-based Medicine. Cambridge, UK:
Cambridge University Press; 2004.
9. Katcher BS. Medline: A Guide to Effective Searching in
PubMed and Other Interfaces. 2nd ed. San Francisco, CA:
Ashbury Press; 2006.
10. Detwiler SM. Super Searchers on Health & Medicine: The
Online Secrets of Top Health & Medical Researchers. Medford,
NJ: Cyberage Books; 2000.
11. Liu JH. Guides to an efficient search for the best evidence.
Seminars in Reproductive Medicine. 2003; 21(1):5–8.
60 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
12. Murphy LS, Reinsch S, Najm WI, et al. Searching biomedi-
cal databases on complementary medicine: the use of con-
trolled vocabulary among authors, indexers, and investiga-
tors. BMC Complementary & Alternative Medicine. 2003; 3:3.
13. Dixon RA, Munro JF, Silcocks PB. The Evidence Based Medi-
cine Workbook: Critical Appraisal for Clinical Problem Solv-
ing. Oxford, UK: Butterworth – Heinemann; 1997.
14. Glasziou P, Del Mar C, Salisbury J. Evidence-based Medi-
cine Workbook: Finding and Applying the Best Research Evi-
dence to Improve Patient Care. London, UK: BMJ Publish-
ing Group; 2003.
15. Guyatt G, Rennie D, eds. User’s Guides to the Medical Lit-
erature. Chicago, IL: AMA Press; 2002.
16. University of Kentucky College of Pharmacy Drug Infor-
mation. Assessment of quality of internet sites. Available
at: www.mc.uky.edu/pharmacy/dic/assessment_sites.
html. Accessed October 4, 2007.
17. Heneghan C, Badenoch D. Evidence-based Medicine Toolkit.
Oxford, UK: Blackwell Publishing Limited; 2006.
18. Levi M. Formulating Clinical Questions. In: McGovern
DPB, Summerskill WSM, Valori RM, et al. Key Topics in
Evidence-Based Medicine. Oxford, UK: BIOS Scientific Pub-
lishers Limited; 2001.
19. Pace HA. Evidence-based practice guidelines: Pearls for
the clinician. Paper presented at: American Society of
Health-System Pharmacists Annual Midyear Clinical Meet-
ing; December 4, 2006; Anaheim, CA.
Chapter 5
Evaluate
Literature
Morgan L. Sperry & Patrick J. Bryant
Step 3 – Evaluate Literature
Introduction
Multiple Item Checklists
Ten Major Considerations
Summary
References
Step 3 – Evaluate Literature
After the clinical question has been defined and all pertinent in-
formation in answering it has been retrieved, the practitioner will
perform a critical literature evaluation on all primary material
collected. This third step in the 5-Step Evidence-Based Medicine
Process is vital to determine any major limitations in a study and
the impact they may have on the results and/or primary out-
comes of the study.
Introduction
Most practitioners have experience in critical literature evalua-
tion. Multiple item checklists are used to identify and evaluate
important aspects of a study. While these multiple item lists can
be helpful to the evaluator due to the great detail and number of
specific points given to address within a study, they can also be
time consuming and daunting for the practitioner. Health care
professionals have a limited amount of time when deciding what
articles to read and critically evaluate.
61
62 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
The purpose of this chapter is to reduce the burden of critical
literature evaluation placed on the practitioner by introducing
the Ten Major Considerations. The Ten Major Considerations
are used to identify strengths and limitations in a study that have
the greatest impact on the results/conclusions. Applying these
considerations makes step three of the 5-Step Evidence-Based
Medicine Process more efficient and user friendly for the busy
practitioner.
Multiple Item Checklists
Often multiple item checklists are used as a teaching aid for stu-
dents who are evaluating literature. An example of a multiple
item checklist is provided at the end of this chapter. While these
lengthy lists serve a purpose when orienting students to the criti-
cal literature evaluation process, they can be a barrier to the busy
practitioner trying to assess studies in time-sensitive situations.
Many items on multiple item checklists do not have a major im-
pact on the study’s overall validity nor do they shed much light
on the reliability of the evidence provided by a study. In narrow-
ing down these extensive checklists to the Ten Major Consider-
ations, practitioners will find critical literature evaluation much
more functional (see the table in Figur
Figuree 5.1 and also the “EBM
Tool Kit” at the end of this book). The amount of time needed to
thoroughly evaluate the integrity of an article is greatly reduced
by using the Ten Major Considerations.
Ten Major Considerations
Once the Ten Major Considerations have been integrated into
the practitioner’s thought process, reading articles and determin-
ing their relevance become different. Major limitations that could
have a significant impact on the study’s integrity are taken into
account, thus allowing the practitioner to recognize potential
problems with the overall outcome of the study. By evaluating
articles from this perspective, the practitioner is able to quickly
identify the overall quality of evidence as well as which articles
are worth reading.
Chapter 55: Evaluate Literature 63
Ten Major Considerations Strength Limitation
Strength
Power set/met?
Dosage/treatment regimen appropriate?
Length of study appropriate to show effect?
Inclusion criteria adequate?
Exclusion criteria adequate?
Blinding present?
Randomization resulted in similar groups?
Biostatistical tests appropriate for type of
data analyzed?
Measurement(s) standard/validated/accepted
practice?
Author's conclusions are supported by the
results?
Figur
iguree 5.1: Ten Major Considerations
The Ten Major Considerations provide the practitioner with
a concise and quick method to critically evaluate literature, a
method that is much less daunting and time consuming com-
pared to using a multiple item checklist. The Ten Major Consid-
erations are presented in the table above and in detail below.
1. Is power set and/or met? Power is the ability of a study
to detect statistically significant differences between treat-
ment groups, when such a difference truly does exist. Once
a study is verified as a randomized controlled trial, the
practitioner can establish whether or not power was met.
To determine this, the description for power should be
found in the statistical section of most studies. When
deciding whether or not a particular study met power,
the practitioner should ask the following questions:
What is power set at?
How many patients are needed to meet power?
64 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
What type of analysis is performed on the primary out-
come measurements—per protocol or intent-to-treat?
Does the number of patients used in the analysis meet
the sample size required to meet power? If so, one
should feel confident that enough patients are in-
cluded to show a difference if one exists.
With practice, the above questions involving power should
become “second nature” to the practitioner when look-
ing at a study.
Example 5.1. Power Is Met
Power
In a study conducted by Dahl et al.,1 total sample
size was based on estimated differences in average
asthma exacerbation rate between two different
treatments. “To detect a difference of 20% in the
relative risk of exacerbations between salmeterol/
fluticasone propionate (SFC) and formoterol/
budesonide (FBC) combinations, at a two-sided
α=0.05 significance level with 90% power using
the Normal approximation to the Poisson regres-
sion techniques, 525 patients per group were re-
quired.” Intent-to-treat analyses were used for both
primary and secondary outcome measures, with
the primary outcome being rate of asthma exacer-
bations over a 6-month period. The final intent-to-
treat analysis included 694 patients from the SFC
group and 697 patients from the FBC group.
This example clearly states that 525 patients are
needed in each treatment group to meet power for
the primary outcome. This study undoubtedly met
this requirement with both the SFC and FBC treat-
ment groups, including well over 525 patients in
their final analyses (n=694 and n=697, respectively).
It is also important to determine whether or not a
per protocol or intent-to-treat analysis was completed
when determining power. A per protocol analysis
Chapter 55: Evaluate Literature 65
requires knowing the number of patients that com-
pleted the study. An intent-to-treat analysis requires
knowing the number of patients randomized into
the study groups. Note that a modified intent-to-
treat analysis includes specific criteria that must be
met once a patient is randomized into the study (e.g.,
must have received at least one dose of the study
drug or had at least one follow-up visit).
Understanding the importance of power in a study can be of
great benefit for the practitioner trying to decipher what lit-
erature is worth reading in his or her limited time. In situa-
tions where power is not met or not calculated, the next ques-
tion asked is: “Does a difference exist between treatment
groups?” If a statistically significant difference is noted between
treatment groups, the fact that the study did not meet power
becomes less of a concern. If no statistically significant differ-
ence is found between treatment groups, a possibility exists
that there were not enough people enrolled in the trial to de-
tect a significant difference. This is a concern. Therefore, a
clinician makes best use of time by reading trials that either
did meet power or did not meet power but showed a statis-
tically significant difference (potential type II error). See
Figur
Figuree 5.2 for various power scenarios. For additional in-
formation on power, see Chapter 2: Basics for Interpretation.
Example 5.2. Power Is Not Met
Power
In a trial performed by Gonzales et al.,2 all study
participants were randomized and all efficacy data
was compared via intent-to-treat analysis. “A
sample size of 335 participants per group was es-
timated as providing 90% power for a 2-tailed x2
test with α=0.05 for the comparison between
varenicline and bupropion SR for the 4-week con-
tinuous abstinence rate based on an odds ratio
66 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Statistically significant difference between groups detected?
Yes No
Power not as great Power analysis performed?
of a concern
Yes No
Is sample size large
enough to represent the
general population? Power Met? Unable to assess if study
could detect a difference
if one existed (Major
Yes No Limitation)
Study powered to detect a Study not powered to detect a
difference (Major Strength) difference (Major Limitation)
Figur
iguree 5.2: Power Scenarios
(OR) of 1.72 vs a bupropion SR response rate of
28.6%.” Four-week continuous rates of abstinence
between trial weeks 9 through 12 were confirmed
via exhaled carbon monoxide. Varenicline (n=352)
proved superior to placebo (n=344) (95% CI,
2.70–5.50; P<.001) and buproprion SR (n=329)
(95% CI, 1.40–2.68; P<.001).
This example states that a sample size of 335
patients per group is needed to have 90% power.
This trial failed to meet this goal with the
buproprion SR group falling just short of the 335
patients required (n=329). While this study did not
meet power, it is less of a concern because a statis-
tically significant difference was seen in regards to
the results. Varenicline proved to be superior to
placebo and buproprion SR (P<0.001). In this case
it would have been better for the trial to have met
power, but since a statistically significant difference
was still seen it was less of a concern and the po-
tential for type II error was diminished.
Chapter 55: Evaluate Literature 67
2. Ar
Aree dosages and tr eatment rregimens
treatment egimens appr opriate? It is
appropriate?
important to look at the dosage and/or treatment regimen
used for each treatment group in a study. A potential major
limitation can exist if a dosage and/or treatment regimen
being used is not typically what is seen for the specific in-
dication being addressed. This can help the practitioner
recognize if sub- or supra-therapeutic dosing may have an
effect on study results. It is also important to consider
whether or not equivalent doses of study drugs are being
compared. For instance, in a comparative trial evaluating
cholesterol-lowering agents, one would not want
simvastatin 80 mg to be compared to atorvastatin 10 mg.
This comparison could potentially provide an advantage
for one drug over the other and confound the results.
Example 5.3. Doses and R egimens Ar
Regimens Aree
opriate
Appropriate
Appr
In a study conducted by Manzoni et al.,3 very low-
birth-weight infants received fluconazole for pre-
vention of fungal infections. For 2 weeks, infants
were given 3 or 6 mg of fluconazole every third
day; after the 2-week period, this became every
other day. Normal saline was given on this same
timetable to the placebo group. The study drug
was administered intravenously through a cath-
eter, if in use, or an orogastric tube.
In this example, a determination needed to be
made in regards to appropriate dosing in the pe-
diatric population. The main concern is safety of
the dosage given in this study. Once safety is es-
tablished, it is then appropriate to determine
whether the dose of fluconazole used is similar to
doses previously used for this indication. The Lexi-
Comp Pediatric Dosage Handbook is a great resource
for this example and helps the practitioner easily
establish if the dosage being used in the study is
68 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
suitable. The dosage in the above excerpt is a good
example of a trial using an appropriate dosage regi-
men.
It is not always clear whether a dosage and/or regi-
men is appropriate when looking at a study that is inves-
tigating a non-FDA approved indication. Compendia such
as Lexi-Comp and Clinical Pharmacology are helpful re-
sources that refer to some non-FDA approved indications
and doses. The safety of the dosage and/or treatment regi-
men is the primary concern. This can often be determined
by reviewing other approved indications for the treatment
in question to determine if dosages being used in the trial
fall within these approved dosage ranges. In addition,
some literature may address dosing for the particular non-
FDA approved indication being studied.
Example 5.4. Doses and R egimens for
Regimens
a Non-FD
Non-FDAA Appr oved Indication
Approved
Legro et al.4 performed a trial examining clomi-
phene and metformin’s efficacy in infertility asso-
ciated with polycystic ovarian syndrome (PCOS).
Metformin and clomiphene were begun concur-
rently upon study initiation. Patients received
metformin 500 mg and clomiphene 50 mg,
metformin 500 mg and matching clomiphene pla-
cebo, matching metformin placebo and clomi-
phene 50 mg, or just placebo. Subjects receiving
metformin 500 mg increased their dosage to two
tablets twice daily (maximum dose of four tab-
lets) depending on their tolerability of the drug.
Subjects randomized to clomiphene were given
one 50-mg clomiphene tablet to be taken for 5
days beginning on the third day of menses. Docu-
mentation of adequate ovulation determined
Chapter 55: Evaluate Literature 69
whether clomiphene dosage should be main-
tained. A dosage increase of one additional 50-
mg tablet per day was made in patients with no
response or minimal response to clomiphene.
In cases where a study is evaluating a non-
FDA approved indication like the above example
for metformin and polycystic ovarian syndrome
(PCOS), the first concern should be for the safety
of the particular dosage regimen in patients. Lexi-
Comp and Clinical Pharmacology are resources that
may include recommended dosing for non-FDA
approved indications. If unable to confirm appro-
priate dosing using these resources, the
practitioner’s next step is to ensure dosing is within
appropriate ranges seen in indications in which
the medication has been approved. In this case,
the non-FDA approved dose for metformin in
PCOS is listed in Clinical Pharmacology as 500 mg
three times daily. While the above study uses a
higher dosage for PCOS than reported in Clinical
Pharmacology (two 500-mg tablets twice daily), it
is still considered appropriate because it is not out
of range when looking at an FDA-approved indi-
cation for metformin such as diabetes.
3. Is the length of the study appr opriate to show ef
appropriate fect?
effect?
A study conducted for an inadequate length of time can
result in no treatment effect seen when there really is po-
tential for an effect. In most situations, the concern will
be whether or not the trial was conducted for a long
enough period of time. For instance, some antidepres-
sant medications such as selective serotonin reuptake in-
hibitors (SSRIs) require 4–6 weeks to see any effect or
improvement in patients. A study involving these agents
only lasting 3 weeks may not be an adequate amount of
time to show any significant differences in efficacy.
70 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
4. Ar
Aree inclusion criteria adequate? Inclusion criteria are
developed primarily to ensure the correct patient popu-
lation is used to determine efficacy. To participate in any
well-designed study, subjects must meet certain criteria
before being approved for inclusion in a trial. Investiga-
tors identify specific parameters such as age, disease state
and/or stage of disease state, sex, treatment history, and
other medical conditions that may influence trial results.
When inclusion criteria are not properly defined, the
possibility exists that a study may be using patients not
representative of the population that the investigators in-
tended to treat. Correctly selecting the inclusion criteria
for a trial can have a large impact on whether or not the
study is clinically relevant to the question being asked.
Choosing to include patients outside the target disease
state or whose demographics are not appropriate for the
desired population may have an impact on the results of
the study. In these cases, clinical judgment must be used
to determine the extent of the effect.
Example 5.5. Inclusion Criteria
Tfelt-Hansen and associates5 conducted a study
comparing sumatriptan versus rizatriptan in acute
treatment of migraine. Patients who met proper
criteria for migraine with or without aura as de-
termined by the International Headache Society
(IHS) were enrolled in the trial. Subjects were re-
quired to be in good health and to have had at
least a 6-month history of migraines, experienc-
ing between one and eight attacks per month.
Enrolled subjects were both male and female aged
18–65 years.
When determining whether or not inclusion
criteria are appropriate, it is important to keep in
mind why criteria are developed in the first place.
Trial investigators must ensure that the correct
Chapter 55: Evaluate Literature 71
patient population is used to determine efficacy,
otherwise a study may be evaluating a population
that is not representative of the one they set out
to treat. In the example shown above, it was the
investigator’s intent to compare two migraine
medications within the same class and the effect
they had on patients with acute migraine. The in-
clusion criteria developed in this example resulted
in a patient population appropriate for the study.
Practitioners who are evaluating inclusion crite-
ria should always ask whether the patient popu-
lation enrolled is representative of the population
they intend to treat.
When evaluating a study for adequate inclusion cri-
teria, the practitioner should also identify any inclusion
criteria not mentioned that would be important to the
specific trial. If certain inclusion criteria are identified as
absent, the practitioner must assess whether or not there
is a potential effect on the results due to this deficit.
5. Ar
Aree exclusion criteria adequate? Patient safety is the pri-
mary focus when establishing exclusion criteria. This is
the investigator’s opportunity to exclude any patients who
may be at risk for increased adverse events and other safety
issues by participating in the trial. Just as investigators de-
termine specific parameters or guidelines to establish those
patients to be included in a trial, parameters are also estab-
lished for patients who will be excluded because of safety
reasons. Factors such as co-morbid conditions and con-
current medications that have the possibility of increasing
the risk of adverse events are taken into consideration.
Exclusion criteria not only ensure safety in a study
but guard against inclusion of patients who could poten-
tially decrease one’s ability to determine if results are due
to the study treatment or another factor. Below are a few
examples of these potential factors:
72 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Previous exposure to the study treatment/similar treat-
ment within 6 months of the study
Any current medication use that could bias the results
Certain co-morbid conditions and/or disease states
Previous procedures affecting the study drug or dis-
ease state in question
It is also important to identify instances when exclusion
criteria are too restrictive, thus creating a study less relevant
to the specific patient population requiring treatment.
Example 5.6. Ex clusion Criteria
Exclusion
A multi-center trial to assess low-dose orlistat ef-
fects on body weight of mild to moderately over-
weight individuals was conducted by Anderson et
al. Subjects were excluded if they had any of the
following: prior weight loss surgery, prior weight
loss of 3 kg or more in the past 3 months, use of
any medications that have an effect on weight loss
within the past 6 months, bulimia and/or laxative
abuse, untreated thyroid disease, history within the
past 6 months of CABG or angioplasty procedure
with cardiovascular disease, diabetes currently con-
trolled by medication, recent smoking cessation
within last 6 months, and use of any medications
that interact with orlistat. Pregnant or breastfeeding
women were not included.
When determining whether a study has selected
appropriate exclusion criteria, emphasis is placed
on patient safety. It is the investigator’s job to make
sure all patients who potentially could be safety risks
are excluded from the trial. Many times this means
learning more about the medication being studied
and its adverse effects. The above exclusion criteria
in the orlistat trial adequately addressed safety con-
cerns such as drug interactions, pregnancy, and
cardiovascular considerations.
Chapter 55: Evaluate Literature 73
The above trial also accounted for concurrent
medications and/or underlying disease states that
could have impacted the results. For instance, in-
vestigators excluded patients with untreated thy-
roid disease such as hypothyroidism or any use of
pharmacological agents within the last 6 months
affecting body weight. With the exclusion of these
factors, the investigators decreased the possibility
that results of the trial would show anything but
the effect orlistat had on weight loss of patients
enrolled in the study.
Investigators may fail to identify all appropriate ex-
clusion criteria. If certain exclusion criteria were left out,
any potential effect this had on the study results should
be assessed.
Example 5.7. Ex clusion Criteria Ar
Exclusion Aree
Not Adequate
In a study conducted by Kolodony and col-
leagues,7 treatments for acute migraine were com-
pared. Study participants were excluded from the
trial if they used any of the following drugs: pro-
pranolol, methylsergide, and monoamine oxidase
inhibitors. The use of standard antimigraine pro-
phylactic medications was allowed except for the
use of NSAIDs, daily analgesics, or propranolol.
Pregnant or lactating women were not eligible for
inclusion into the study.
The above study defined many exclusion cri-
teria that were appropriate for ensuring the safety
of trial participants when using the triptan medi-
cations. Unfortunately, the authors neglected one
major safety issue with the triptan medications:
cardiovascular concerns. Serotonin receptor ago-
74 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
nists, such as the triptans, may increase the po-
tential for cardiovascular side effects; this should
be addressed within the exclusion criteria. When
evaluating the adequacy of exclusion criteria, the
practitioner should be familiar with all safety con-
cerns of the particular medication or treatment
being studied.
6. Is the study blinded? An adequately blinded study guards
against investigator and subject bias. Typically, one would
like to see double-blinded studies because they attempt
to prevent the largest degree of investigator or patient bias
in a trial. In single-blinded studies (where either the in-
vestigator or patient is not blinded to the intervention),
there is a greater chance for bias to confound the results.
The exact method of blinding is seldom explained in
the article. Rather, a statement that the study was blinded
is usually seen. The practitioner should question whether
there were any potential barriers to successfully blinding a
particular drug. For instance, a peculiar smell or taste char-
acteristic of the study drug should also be reproduced for
the control(s) to maintain blinding. That smell or taste in
some cases can be masked for the study drug. In either
situation, an explanation of how this was handled should
be provided. If there is no explanation, the practitioner
must assume the issue was not addressed. Blinding is con-
sidered a major limitation when this occurs; however, the
practitioner still needs to assess whether that major limita-
tion had any effect on the overall results/conclusions.
Example 5.8. Blinding
A double-blind, placebo-controlled trial in patients
with pulmonary hypertension performed by Galie
et al. 8 used a stratified central-randomization
scheme to allocate patients to their four respec-
Chapter 55: Evaluate Literature 75
tive treatment groups (sildenafil 20, 40, or 80 mg
or placebo). Dose escalation took place for the first
7 days of the study for those patients assigned to
the 80-mg treatment group. They received 40 mg
three times daily for 7 days and then moved up to
80 mg three times daily. Dummy dose escalation
was also performed for the other three treatment
groups during the first 7 days.
Although the trial described above was double
blinded and took measures to conceal dose escala-
tion in the sildenafil 80-mg treatment group, there
is still potential for blinding to be flawed. Sildenafil’s
side effect profile includes incidence of penile erec-
tion in male patients, which could potentially cause
patients to become unblinded to their respective
treatment groups. The authors do not address this
potential weakness in the blinding of the study.
7. Did randomization rresult
esult in similar gr oups? When ran-
groups?
domization is successful, each patient has an equal chance
of being assigned to one treatment group versus the other(s).
Even more critical is that the trial ends up with very simi-
lar groups demographically following randomization. If a
significant disparity is found between treatment groups,
there is concern about whether the results observed are
due to the study drug or a difference that was present be-
tween groups from the start. For instance, in a clinical trial
evaluating the efficacy of an asthma medication, it is im-
portant that a similar number of smokers are randomized
to each treatment group. If groups are dissimilar, the re-
sults seen may not accurately depict the medication’s effi-
cacy and may potentially be due to a lower or higher num-
ber of smokers being randomized to a particular group.
The demographics table in a study is useful for con-
firming similar treatment groups. When dissimilarities are
found, it is important to question what impact or clinical
76 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
relevance the dissimilarity could have had on the results.
For instance, disease states often have many levels of se-
verity. If one treatment group has more severe patients
than the other after randomization, the drug’s efficacy may
be falsely reported as reduced in that group compared to
the group with less severe patients. The practitioner must
also be aware of any additional demographic information
that should have been collected or discussed but was not.
If this occurs, it is important to question what impact
this discrepancy may have had on the results.
Example 5.9. Similar Gr oups af
Groups ter
after
Randomization and Statistics
A modified table below illustrates baseline char-
acteristics seen in a trial evaluating the effects on
fracture rate of continuing or stopping alendronate
after 5 years of treatment by Black et al.:9
(5 mg/d) (10 mg/d)
(n=437) (n=329) (n=333)
Characteristics Placebo Alendr onate Alendr
Alendronate onate P V
Alendronate alue
Value
Body mass index, 25.8 25.7 25.9 .05
mean (SD) (4.3) (4.2) (4.5)
Race
White 421 322 327 .22
(96.3) (97.9) (98.2)
Other 16 7 6 .22
(3.7) (2.1) (1.8)
Walk for exercise 244 189 203 .40
(57.0) (58.7) (61.9)
Fall in last 12 mo. 105 80 71
(24.2) (24.4) (21.6)
History of clinical 260 196 204 .86
fracture (>45 (59.5) (59.6) (61.3)
years old)
Current 341 275 262 .13
alendronate use (78.0) (83.6) (78.7)
Chapter 55: Evaluate Literature 77
The above example demonstrates how some tri-
als display demographic data. Statistics were run
on the demographic data above, helping the prac-
titioner to detect any significant differences seen
in baseline characteristics after randomization.
When studies provide statistical information on
baseline data, it makes identifying significant dif-
ferences much easier. For instance, if p-values were
not provided, one might question whether or not
a significant difference was seen. No significant
differences were seen in the above example. If a
significant difference had been seen, the practi-
tioner should question what impact the difference
would have had on the results of the trial.
Example 5.10. Similar Gr oups af
Groups ter
after
Randomization and No Demographic
Table
In a study conducted by Anderson et al.,6 “the
baseline demographics of the placebo and orlistat
groups were similar. Of the 378 subjects, 94.4%
were female (n=357), 89.2% were white (n=337),
average (+SD) age was 46.2 + 11.41 years, baseline
weight was 72.8 + 6.97 kg, and BMI was 26.8 +
0.96 kg/m2.”
Not all studies will address the similarity of
demographics among treatment groups after ran-
domization in the same way. For instance, the
above example is from a trial that just briefly men-
tioned groups were similar in the text of the ar-
ticle. No chart or statistics were given. In these
cases, the practitioner must decide whether this
major consideration was adequately addressed and
what impact may be seen on the results.
78 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Statistical tests are not always performed on demographic
information in a trial (see Example 5.10 5.10). This means the
practitioner is not always provided with information about
whether statistically significant differences are seen between
each group’s demographic characteristics. In these instances,
a judgment call must be made by the practitioner to deter-
mine if any differences exist that may impact the results.
Example 5.11. Similar Gr oups af
Groups ter
after
Randomization and No Statistics
An open-label, parallel group, randomized con-
trolled trial by Farmer et al.10 evaluated the im-
pact of self monitoring of blood glucose in pa-
tients with non-insulin treated diabetes:
Less Intensive Moree Intensive
Mor
Self-Monitoring Self-Monitoring
Characteristics Control Gr
Control oup
Group Group
Group Group
Group
Mean (SD) age 66.3 (10.2) 65.2 (10.6) 65.5 (9.9)
(years)
Men 85 (55.9) 88 (58.7) 87 (57.6)
Median 3 (2–6) 3 (2–7) 3 (2–6)
(interquartile
range) duration
(years) of diabetes
Treatment
Diet only 44 (28.9) 39 (26.0) 41 (27.2)
Monotherapy 57 (37.5) 58 (38.7) 58 (38.4)
Combined 51 (33.6) 53 (35.3) 52 (34.4)
oral therapy
Use of blood
glucose meter
Not using 104 (68.4) 110 (73.3) 102 (67.5)
Using once 48 (31.6) 40 (26.7) 49 (32.5)
weekly or less
Mean (SD) 7.49 (1.09) 7.41 (1.02) 7.53 (1.12)
hemoglobin A1c
(%)
Chapter 55: Evaluate Literature 79
Although a chart was provided in this study to il-
lustrate baseline characteristics, the table was not
analyzed for statistically significant differences seen.
In these instances, the practitioner must use clini-
cal judgment to determine if any demographic in-
formation appears to be significantly different. If
something is identified, the next step is question-
ing what effect that difference will have on results.
When analyzing data using a per protocol analysis,
one should ensure that groups are similar at the end of
the study. A per protocol analysis utilizes the results seen
in each of the treatment groups at the completion of a
study. Patients withdraw from studies for various reasons,
which can result in different groups at the end of the trial.
When evaluating a study using a per protocol analysis,
the evaluator should attempt to determine if the groups
are similar at completion of the trial. When a difference
between groups occurs at the end of a study, it is impor-
tant to question what impact or clinical relevance the dis-
similarity may have on results.
Finally, it is important to remember that even a well
planned and adequately conducted randomization can fail
and result in dissimilar groups. In this instance, the practi-
tioner evaluating the study must determine if these differ-
ences have any significant impact on the results.
8. Ar
Aree appr opriate statistical tests used for type(s) of data
appropriate
analyzed? The reader must determine if the appropriate
statistical test was used for the type of data analyzed. This
particular consideration tends to create the most diffi-
culty for the practitioner. Using the incorrect statistical
test for the type of data analyzed can have serious nega-
tive impact on the results, making them impossible to
interpret accurately. When the incorrect statistical test is
used for the type of data analyzed, the results should not
be trusted unless the authors have provided an adequate
80 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
explanation why the tests in question were used. Luckily
for the practitioner, this determination does not have to
be such a tedious and puzzling process. When one is
equipped with the right tools, identifying the correct sta-
tistical test in a trial can be straightforward. A good un-
derstanding of the different types of data (nominal, ordi-
nal, interval, and ratio) used in trials and access to an
appropriate statistical test reference is all that is neces-
sary. A chart detailing when certain statistical tests should
or should not be used is found in Chapter 2: Basics for
Interpretation, Figure 2.13.
9. Ar
Aree evaluation measur ements standar
measurements d, validated, or
standard,
accepted practice? When choosing how to accurately
measure the primary outcome of a study, the investiga-
tors should select evaluation measurements that are vali-
dated or accepted in practice. In addition, it is important
to verify that the outcome measure being used is relevant
to the disease state in question. For instance, weight loss
should not be used as a primary outcome measure in a
trial investigating the efficacy of an oral diabetic agent in
lowering blood glucose levels.
Example 5.12. Appr opriate Evaluation
Appropriate
Tool
In a trial performed by Sastry and associates,11 the
efficacy of sildenafil versus placebo for primary
pulmonary hypertension was studied. The
Naughton protocol was used to help evaluate the
primary endpoint of the study, which was “change
in exercise time on treadmill.”
It is important to establish the appropriate-
ness of both the primary outcome of a study as
well as the evaluation tool used to measure that
primary outcome. Determining whether an out-
come measure and/or evaluation tool is validated
Chapter 55: Evaluate Literature 81
or accepted in practice can be done in a variety of
different ways. For instance, the above example
uses “change in exercise time on treadmill” as the
primary outcome measure and the Naughton pro-
tocol as the evaluation tool. A quick literature
search confirms that both this outcome measure
and evaluation tool are frequently used when look-
ing at the disease state of primary pulmonary hy-
pertension. Also, the Naughton protocol is de-
scribed in many clinical exercise physiology text-
books as being an approved method for treadmill
tests. Therefore, both the outcome measure and tool
are validated and accepted methods in practice.
Even if the results of a study are statistically signifi-
cant, they are questionable and potentially not applicable
if an inappropriate evaluation method is used to measure
the primary outcome.
If you are unfamiliar with an outcome measure or test
being used in a particular study, further investigation is
warranted. When the measurement or test in question is
referenced, the practitioner can go to the reference cited
to determine if the measurement or test has been vali-
dated and/or is an accepted practice. In cases where the
evaluation tool in question has not been referenced, the
following questions may be asked when determining the
appropriateness of the outcome measure or test.
Do studies looking at this same outcome use this par-
ticular evaluation tool?
Is this method or test validated?
Does the evaluation method being used measure
something that is clinically relevant to the disease state
or medication being studied?
Is this a new trend being used to evaluate this specific
outcome?
82 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
10. Ar
Aree author’s conclusions suppor ted by the rresults?
supported esults? Fi-
nally, results and/or primary outcomes seen in studies
must support the conclusions made by the authors of
those articles. There should be concern when conclusions
are drawn by study investigators that do not seem to co-
incide with the results reported in a trial. This kind of
disparity between the results of a study and the conclu-
sion may indicate investigator bias and should be ques-
tioned.
Summary
While multiple item checklists can be a very detailed and specific
tool for introducing critical literature evaluation, it is unrealistic
to believe this is the most efficient way to evaluate literature for
the busy practitioner. Identification of the Ten Major Consider-
ations and how they impact the results allows the practitioner to
not only evaluate literature effectively but also reduce the amount
of time spent performing this task. Integration of these consider-
ations into the practitioner’s thought process can change the ap-
proach by which articles are read and evaluated. This will also
aid the practitioner in determining what articles are worthy of
his or her limited time.
References
1. Dahl R, Chuchalin A, Gor D, et al. EXCEL: A randomized
trial comparing salmeterol/fluticasone propionate and
formoterol/budesonide combinations in adults with per-
sistent asthma. Respiratory Medicine. 2006; 100:1152–
1162.
2. Gonzales D, Rennard SI, Nides M, et al. Varenicline, an
α4β2 nicotinic acetylcholine receptor partial agonist, vs
sustained-release bupropion and placebo for smoking
cessation, a randomized controlled trial. JAMA. 2006;
296:47–55.
Chapter 55: Evaluate Literature 83
3. Manzoni P, Stolfi I, Pugni L, et al. A multicenter, random-
ized trial of prophylactic fluconazole in preterm neonates.
N Engl J Med. 2007; 356:2483–2495.
4. Legro RS, Barnhart HX, Schlaff WD, et al. Clomiphene,
metformin, or both for infertility in the polycystic ovary
syndrome. N Engl J Med. 2007; 356:551–566.
5. Tfelt-Hansen P, Teall J, Rodriguez F, et al. Oral rizatriptan
versus oral sumatriptan: A direct comparative study in
the acute treatment of migraine. Headache. 1998; 38:748–
755.
6. Anderson J, Schwartz S, Hauptman J, et al. Low-dose
orlistat effects on body weight of mildly to moderately
overweight individuals: a 16 week, double-blind, placebo-
controlled trial. Ann Pharmacother. 2006; 40:1717–1723.
7. Kolodony A, Polis A, Battisti WP, et al. Comparison of
rizatriptan 5 mg and 10 mg tablets and sumatriptan 25
mg and 50 mg tablets. Cephalalgia. 2004; 24:540–546.
8. Galie N, Ghofrani H, Torbicki A, et al. Sildenafil citrate
therapy for pulmonary arterial hypertension. N Engl J Med.
2005; 353:2148–2157.
9. Black D, Schwartz A, Ensrud K, et al. Effects of continu-
ing or stopping alendronate after 5 years of treatment.
The fracture intervention trial long-term extension
(FLEX): a randomized trial. JAMA. 2006; 296:2927–2938.
10. Farmer A, Wade A, Goyder E, et al. Impact of self moni-
toring of blood glucose in the management of patients
with non-insulin treated diabetes: open parallel group
randomized trial. BMJ. 2007; 335:132–139.
11. Sastry BK, Narasimhan C, Reddy NK, et al. Clinical effi-
cacy of sildenafil in primary pulmonary hypertension: a
randomized, placebo-controlled, double-blind, crossover
study. J Am Coll Cardiol. 2004; 43:1149–1153.
84 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Evaluation of Primar
Primar
rimaryy Literature: Multi-item Checklist
Literature:
Ar ticle T
Article itle:
Title:
Checklist Item Y/N S/W Justification
1. Is the journal peer-reviewed?
2. Is title biased?
3. Are investigators considered reliable and was
study conducted in a reputable medical center or
university teaching hospital? If not, is location
adequate for application of good scientific
experimental method?
4. Are objectives and/or hypothesis (i.e., purpose)
clearly defined?
5. Were inclusion criteria appropriate for the
purposes of the study?
6. Were exclusion criteria appropriate for the
purposes of the study?
7. In addition to inclusion/exclusion criteria, was
adequate pertinent patient information provided
(e.g., disease severity, demographics, previous
treatment failure, smoking status, lifestyle habits,
etc.)?
8. Were recruitment methods targeted toward
persons likely to be representative of applicable
population?
9. Was trial prospective or retrospective? X X
10. Was a control established?
11. If so, was type of control appropriate?
12. Were study drug and controls allocated randomly?
(If given, what method was used?)
13. Were group characteristics similar after
randomization? If possible to determine, was the
similarity of characteristics maintained throughout
study completion, i.e., after dropouts?
14. Was study blinded and, if so, were blinding
techniques appropriate?
15. Were drug doses and regimens appropriate (e.g.,
within known therapeutic ranges, safe, proper
interval, equally effective doses when active drugs
compared)?
16. Was treatment duration adequate for assessing
effect?
Chapter 55: Evaluate Literature 85
Checklist Item Y/N S/W Justification
17. Were any drugs used concurrently? Was
concurrent drug use discussed?
18. Were outcome measurements validated or
accepted practice?
19. Were steps taken to decrease inter-rater
variability?
20. Did authors discuss any factors or influences that
may have affected results of outcome measures?
Were there additional sources of bias noted?
21. Were there any problems in reporting or accuracy
of results (e.g., did text and table/graph data
agree, any missing data)?
22. Were side effects reported?
23. Were type, severity, and incidence of side effects
reported?
24. Were the number of dropouts reported?
25. Were precise reasons for dropouts discussed?
26. Was a per protocol or an intent-to-treat analysis XXX XXX
used?
27. How many subjects were enrolled? XXX XXX
28. How many subjects were included in final XXX XXX
statistical analysis?
29. Was a power calculation performed?
30. If a power calculation was performed, was power
met at study end?
31. Were all statistical tests used appropriate for types
of data (nominal, ordinal, or interval)?
32. Are conclusions and recommendations consistent
with results obtained?
33. Does article provide a reference list to verify
footnoted citations?
Other str engths/weakness specific to this ar
strengths/weakness ticle
article
1.
2.
3.
Used, with permission, from the University of Missouri–Kansas City School of Pharmacy Drug
Information Center.
Chapter 6
Categorize
Quality of
Evidence
Patrick J. Bryant
Step 4 – Categorize Quality of Evidence
Levels of Evidence—Specific Study Design
Characteristics
Five Questions to Ask
Ten Major Considerations—Key Study Attributes
Summary
References
Step 4 – Categorize Quality of Evidence
After completing the critical evaluation of a specific trial, the prac-
titioner needs to categorize the quality of evidence. This fourth
step is the bridge between performing a critical literature evalua-
tion and developing a recommendation.
Key Categorizing the quality of evidence is the bridge
Idea between performing a critical literature evaluation
and developing a recommendation with justifications.
Quality of evidence is determined by evaluating specific study
design characteristics and key study attributes (see Figur
Figuree 6.1
6.1).
87
88 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
QUALITY OF EVIDENCE =
Specific Study Design Characteristics
(Level of Evidence)
+
Key Study Attributes
(Ten Major Considerations–Strengths/Limitations)
Figur
iguree 6.1: Quality of Evidence Equation
For instance, a randomization scheme, comparison group, and
adequate sample size are specific study design characteristics. In
addition, key attributes that strengthen or weaken the basic study
design are considered.
The quality of evidence equation includes specific
Key study design characteristics and key study
Idea attributes that determine the overall quality of
evidence for a particular study.
These key attributes are represented by the Ten Major Con-
siderations that were previously used to critically evaluate articles
(see Chapter 5: Evaluate Literature). Both components of the qual-
ity of evidence equation (specific study design characteristics and
key study attributes) are required for an accurate categorization
of the quality of evidence.
Levels of Evidence—Specific Study Design Characteristics
A ranking system referred to as levels of evidence is utilized to
categorize studies based on quality of design.
Key The level of evidence for a study categorizes
Idea the overall study design quality.
Chapter 66: Categorize Quality of Evidence 89
Several methods exist to categorize a study’s level of evidence.1–8
The method used in this chapter provides a simple, straightfor-
ward, and logical approach. Originally described by Drs. Deborah
Cook and Gordon Guyatt, this method incorporates a modified
approach.9 The process is based on four specific study design char-
acteristics that assist in categorizing studies by their respective lev-
els of evidence. These specific study design characteristics are
1. Interventional or observational study design
2. Controlled or non-controlled study design
3. Randomized or non-randomized study design
4. Power set/met or not set/met for the study design
Using this method, a study is assigned Levels I through V, with
Level I representing the highest level of study design quality (see
table in Figur
Figuree 6.2
6.2).
Level Quality
of Evidence Study Design Characteristics of Design
Level I Interventional Randomized Controlled *****
Trials with Meeting Power
Level II Interventional Randomized Controlled ****
Trials with Not Meeting Power
Level III Observational Prospective Trials ***
Level IV Observational Retrospective Trials **
Level V Case Studies, Case Series, and Case *
Reports
Figur
iguree 6.2: Levels of Evidence – Study Design Characteristics and Quality
of Study Design
Using the level of evidence ranking system,
Key a study is assigned one of five categories,
Idea Level I through Level V; Level I represents
the highest level of study design quality.
90 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Example 6.1. R elationship Between
Relationship
Study Design Characteristics and
Levels of Evidence
A randomized, controlled trial comparing two
different drugs for use in irritable bowel syndrome
could be
• Level I if powered to show a difference if one
really existed.
• Level II if power is not met or not initially set
to show a difference if one existed.
Studies used to observe changes such as incidence
of constipation versus diarrhea associated with
irritable bowel syndrome without intervention in
a population of patients over time could be
• Level III if a prospective observational study
design is used.
• Level IV if a retrospective observational study
design is used.
A testimonial of how well stress reduction exer-
cises reduce irritable bowel symptoms in a single
patient or series of patients would be considered
• Level V since no randomization scheme, con-
trol group, or set power exists.
The progression from Level I to Level V represents a decrease
in quality of study design and, therefore, reliability of results pro-
vided by that particular trial. As the evidence used to develop a
recommendation decreases in quality (Level I going to Level V),
the overall strength of that recommendation also decreases. In
other words, Level I evidence represents the highest possible level
of study design quality; Level V evidence represents the lowest
level of study design quality. Recommendations are developed
from the highest level of evidence available. When no evidence
exists, recommendations are made based on experience which
can overestimate efficacy and underestimate safety risks.
Chapter 66: Categorize Quality of Evidence 91
Five Questions to Ask
Five questions are asked to determine existence of these specific
study design characteristics within a particular trial. Diagrams
are provided to help illustrate the differences between the five
Figur
levels of evidence (Figures 6.3–6.7
Figures 6.3–6.7).
Five questions are asked and answered to
determine the existence of specific study design
characteristics within a study:
1. Does the study have an intervention or is it
strictly observational?
Key 2. Does the study have a control group?
Idea 3. Does the observational study look forward
or backward in time (prospective or
retrospective)?
4. Is the study randomized?
5. If the study is a randomized and
controlled trial, is power met?
Question #1: Does the study have an inter vention or is it
intervention
strictly obser vational? In other words, are patients being as-
observational?
signed a specific therapy or intervention?
Level I and Level II evidence are represented by trials with
an interventional study design (see Figur
Figuree 6.3
6.3). The in-
vestigators are assigning a specific intervention to each
subject. For instance, a trial to identify the effects of an
anti-diabetic agent would require a group of subjects to
be given the intervention (drug).
Levels III and IV evidence are represented by trials with an
observational study design. In observational trials, investiga-
92 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
tors are merely observing the subjects and no intervention is
being assigned. Subjects are chosen based on presence or
absence of specific characteristics. A trial observing what com-
plications occur to diabetic patients with lack of long-term
glycemic control versus diabetic patients with long-term
glycemic control is considered an observational trial.
Level V evidence is represented by trials with either an
interventional or observational study design.
Note that trials utilizing an interventional study design rep-
resent the higher levels of evidence (Levels I, II). Those trials
utilizing an observational study design represent a lower level of
evidence (Level III, IV), and the trial design representing the lowest
level of evidence is Level V. The progression from Level I to Level
V evidence represents a decrease in quality of trial design and,
therefore, reliability of results provided by that particular trial. A
decrease in quality and reliability of evidence corresponds to a
decrease in overall strength of the recommendation being devel-
oped.
Level I
Interventional
Level II
confidence
Level III
Observational
Level IV
Level V Interventional/Observational
Figur
iguree 6.3: Levels of Evidence – Interventional versus Observational Study
Design
Key The strength of a recommendation is weakened
Idea when evidence of low quality and reliability is used.
Chapter 66: Categorize Quality of Evidence 93
Question #2: Does the study have a contr ol gr
control oup?
group?
Level I and Level II evidence are represented by trials with a
control group (see Figur
Figuree 6.4
6.4). Control groups provide a
comparison to ensure that changes seen in the interven-
tion group are due to something other than chance.
Level III and Level IV evidence are also represented by trials
with a control group. This control group can be either
prospective or retrospective in nature. As with
interventional trials, the control group serves as a way to
ensure that changes seen in the study group are due to
something other than chance.
Level V evidence is represented by trials without a control
group. Without a control group, there is no way to con-
firm that changes seen in a subject or group of subjects
are due to something other than chance. Essentially, Level
V trials are equivalent to personal testimonials. Level V
trials typically consist of a single-case study, case series,
and/or case report.
Note that trials containing a control group represent the higher
levels of evidence (Levels I through IV). Those trials containing
no control group represent the lowest level of evidence (Level V).
The progression from Level I to Level V evidence represents a
decrease in the quality of the trial design and, therefore, reliabil-
ity of the results provided by that particular trial. Without high
quality and reliabile evidence, the strength of the recommenda-
tion is compromised.
Control Group
Level I
Control Group Interventional
Level II
confidence
Control Group
Level III
Observational
Level IV
Control Group
Level V Interventional/Observational
No Control Group
Figur
iguree 6.4: Levels of Evidence – Control Group versus No Control Group
Study Design
94 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Question #3: Does the obser vational study look for
observational war
forwar
wardd or
backwar
backward d in time (pr ospective or rretr
(prospective etrospective)?
etrospective)?
Level III evidence is represented by observational trials that
look forward in time using prospective cohort groups of
subjects (see Figur
Figuree 6.5
6.5). In prospective observational
trials, the investigators define the disease and the pre-
dicted variables prior to the onset of disease. Subjects are
chosen based on specific characteristics (e.g., smoking)
and followed forward in time to observe the development
of a specific outcome (e.g., lung cancer). For a more de-
tailed description of prospective observational trials, see
Chapter 2: Basics for Interpretation.
Example 6.2. Level III P
Prrospective
Obser vational T
Observational rial
Trial
Level III evidence is represented by a trial such as
the Harvard Nurse’s Health Study II that is con-
sidered one of the most significant studies con-
ducted on the health of women. This study has
been following nurses and the progression of their
health since 1989. An example of the results in-
cludes the long-term effect of moderate alcohol
consumption on the nurse’s cognitive function.
In this analysis, cognitive function was monitored
over time in nurses consuming moderate quanti-
ties of alcohol. A similar group of nurses who were
not consuming alcohol were followed simulta-
neously over the same time period and represent
the prospective cohort comparison group.
Level IV evidence is represented by observational trials that
look back in time using retrospective cohort or case con-
trol groups. For instance, investigators will examine the
data for a group of subjects with a diagnosis of lung can-
cer and examine smoking history of the group to deter-
mine if an association exists between smoking and devel-
opment of lung cancer. For a more detailed description
Chapter 66: Categorize Quality of Evidence 95
of retrospective observational trials, see Chapter 2: Basics
for Interpretation.
Example 6.3. Level IV R etr
Retr ospective
etrospective
Obser vational T
Observational rial
Trial
An observational study is designed to examine the
differences in lifestyle between patients that have
developed esophageal cancer versus those who
have not developed this disease. The investiga-
tors randomly select charts from an institution’s
medical records of patients who developed esoph-
ageal cancer. Various characteristics such as
lifestyle are identified that may have led to the
development of the esophageal cancer such as a
consistent diet of extremely spicy foods.
Note that observational trials using a prospective design repre-
sent a higher level of evidence (Level III) than trials containing a
retrospective design, which represent a lower level of evidence (Level
IV). Trials using a prospective design have less potential for bias
interfering with the results than those with a retrospective de-
sign. The progression from Level III to Level IV evidence repre-
sents a decrease in quality of the trial design and, therefore, reli-
ability of results provided by that particular trial. Only high quality
and reliable evidence can provide a strong recommendation.
Control Group
Level I
Control Group Interventional
Level II
confidence
Control Group
Level III Prospective
Observational
Level IV
Control Group
Retrospective
Level V Interventional/Observational
No Control Group
Figur
iguree 6.5: Levels of Evidence – Prospective versus Retrospective Observa-
tional Study Design
96 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Question #4: Is the study randomized?
Level I and Level II trials utilize a randomization scheme to
assign patients to study groups (see Figur
Figuree 6.6
6.6). Ran-
domization, when done correctly, ensures each subject
has an equal chance of being assigned to either of the
study groups. The result of a trial with a successful ran-
domization scheme has study groups with similar demo-
graphic characteristics following the randomization pro-
cess; differences in an outcome measure between groups
at the end of the study are more likely attributable to the
intervention.
Level III, Level IVIV,, and Level V trials do not utilize a ran-
domization scheme to assign patients into various groups.
Rather, subjects to be entered into the trial can be chosen
at random. For instance, out of a potential subject pool
of 500, the investigators randomly select 300 subjects for
the study. (See Example 6.46.4)
Example 6.4. R andom Selection of
Random
Subjects for Obser vational T
Observational rials
Trials
A prospective observational trial (Level III) ex-
amining the long-term effects of smoking would
include subjects randomly selected from a pool
of subjects with a history of smoking. These pa-
tients are followed forward in time to observe
the development of specific outcomes (e.g., lung
cancer).
Note that trials utilizing a randomization scheme to assign
patients to study groups represent the higher level of evidence
(Level I and Level II). Those trials utilizing no randomization
scheme represent the lower levels of evidence (Levels III, IV,
and V). A successful randomization scheme assures that each
patient has an equal opportunity of being assigned to one group
versus another, and this should result in demographically simi-
Chapter 66: Categorize Quality of Evidence 97
lar groups. Changes noted in the study results can then be at-
tributed to something other than starting out with dissimilar
groups. The progression from Level I to Level V evidence repre-
sents a decrease in quality of the trial design and, therefore,
reliability of results provided by that particular trial. The strength
of the recommendation is decreased when the quality and reli-
ability of the evidence is low.
Control Group
Randomized
Level I
Control Group Interventional
Randomized
Level II
confidence
Control Group
Prospective
Level III Randomly Assigned
Observational
Level IV Control Group
Retrospective
Randomly Assigned
Level V Interventional/Observational
No Control Group
Non-Randomized
Figur
iguree 6.6: Levels of Evidence – Randomized or Non-Randomized Study
Design
Question #5: If the study is an inter ventional randomized
interventional
and controlled trial, is power met?
controlled
Level I evidence is represented by trials where power is set
and met (see Figur
Figuree 6.7 and also the “EBM Tool Kit” at
the end of this book). In these trials, the sample size is
adequate to show a difference between study groups, if a
difference really exists.
Level II evidence is represented by trials where power is set
and not met or power is not set or not mentioned at all. Power
especially becomes important when no statistically signifi-
cant difference is found between the treatment groups. In
this situation, a difference between treatment groups may
have been observed if additional patients were enrolled to
meet power. For this reason, no definitive conclusion can
be made that the groups are similar.
98 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Note that trials where power is set and met represent the
higher level of evidence (Level I). Those trials where power is set
and not met or not mentioned represent the lower level of evi-
dence (Level II). As pointed out in Chapter 2: Basics for Inter-
pretation, power is used to determine an adequate sample size
that will show a difference between treatment groups, if a dif-
ference really exists. The progression from Level I to Level II
evidence represents a decrease in quality of the trial design
and, therefore, reliability of results provided by that particu-
lar trial. Higher quality and reliability of evidence is associ-
ated with an overall increase in strength of the recommenda-
tion.
Please refer to the table in Figur
Figuree 6.8 for a summary of spe-
cific study design characteristics for each level of evidence. In
addition, this table illustrates the degree of confidence associated
with each level of evidence.
Ten Major Considerations—Key Study Attributes
As discussed earlier, the second part of the quality of evidence
equation is the consideration of key study attributes that strengthen
or weaken the basic study design (refer back to Figure 6.1). These
key study attributes are represented by the Ten Major Consider-
ations used to critically evaluate an article (see Chapter 5: Evalu-
ate Literature).
Control Group
Level I Randomized
Power Met Control Group Interventional
Level II Randomized
confidence
Power Not Met Control Group
Prospective
Level III Randomly Assigned
Observational
Level IV Control Group
Retrospective
Randomly Assigned
Level V Interventional/Observational
No Control Group
Non-Randomized
Figur
iguree 6.7: Levels of Evidence – Power Met versus Power Not Met Study
Design
Chapter 66: Categorize Quality of Evidence 99
Inter ventional
Interventional Prospective
Prospective
Level versus Contr ol
Control versus Power
of Obser vational Gr
Observational oup
Group Retrospective Randomized
Retrospective Met
Evidence (I and/or O) (Y es/No)
(Yes/No) (P or R) (Yes/No)
(Yes/No) (Yes/No) Confidence
(Yes/No)
Level I I Y P Y Y +++++
Level II I Y P Y N ++++
Level III O Y P N* - +++
Level IV O Y R N* - ++
Level V I/O N P/R N - +
* Randomly assigned to study, but not to study groups.
Figur
iguree 6.8: Levels of Evidence – Summary of Specific Study Design
Characteristics by Level of Evidence
In addition to the level of evidence, the second
component of quality of the evidence equation is
consideration of key study attributes.
Key These key study attributes are represented by
Idea the Ten Major Considerations used to critically
evaluate an article. Presence of these key
study attributes contributes to strengthening
the quality of the study's design.
If one of the Ten Major Considerations (key study attribute)
is present in the study, that attribute is considered a major strength
(see table in Figur
Figuree 6.9
6.9). However, if that same major consider-
ation or key study attribute is absent or lacking, a major limita-
tion is determined to exist for that study.
100 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Ten Major Considerations Strength Limitation
Strength
Power set/met?
Dosage/treatment regimen appropriate?
Length of study appropriate to show effect?
Inclusion criteria adequate?
Exclusion criteria adequate?
Blinding present?
Randomization resulted in similar groups?
Biostatistical tests appropriate for type of
data analyzed?
Measurement(s) standard/validated/accepted
practice?
Author's conclusions are supported by the
results?
Figur
iguree 6.9: Ten Major Considerations
Level I evidence with minor limitations
represents the highest quality of evidence.
Key
A Level V study—with all Ten Major
Idea Considerations being limitations—is
considered the lowest quality of evidence.
The next question to ask when a major limitation is identi-
fied is whether this limitation appears to have an effect on the
overall study results. If the answer is yes to that question, then the
overall quality of that trial is lessened, even though the level of evi-
dence remains the same.
Chapter 66: Categorize Quality of Evidence 101
Example 6.5. Major Limitation: Impact
versus No Impact on Study R esults
Results
A Level I trial has a major limitation of random-
ization not resulting in similar groups. Upon fur-
ther inspection, this appeared to have an effect on
the results making them questionable. That Level
I trial would have a lesser overall quality of evi-
dence than a Level I trial with randomization iden-
tified as a major limitation and no effect on the
results noted.
Summary
Categorizing quality of the evidence is necessary to determine
the strength of recommendation that can be made based on that
evidence. The quality of evidence equation is a combination of
both specific study design characteristics and key study attributes.
Levels of evidence are determined based on key study design
characteristics such as intervention/observation, controlled/non-
controlled, randomized/non-randomized, and whether power is
set and met by enrolling the required number of subjects.
The better study design provides a higher quality and reli-
ability of evidence (higher level of evidence), making a stronger
recommendation possible. Level I evidence has the highest qual-
ity and reliability of evidence. The progression from Level I to
Level V evidence represents a decrease in the quality and reliabil-
ity of the evidence, and consequently a lower strength of recom-
mendation being developed. Add to this the presence or absence
of key study attributes, and then the overall quality of the evi-
dence can be determined. From this overall quality of evidence
rating, a final recommendation is developed based on the avail-
able evidence.
102 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
References
1. Guyatt GH, Sackett DL, Sinclair JC, et al. Users’ guides to
the medical literature. IX. A method for grading health
care recommendations. Evidence-Based Medicine Work-
ing Group [published erratum appears in JAMA 1996;
275(16):1232]. JAMA. 1995; 274:1800–1804.
2. McGovern DPB, Summerskill WSM, Valori RM, et al. Key
Topics in Evidence-Based Medicine. Oxford, UK: BIOS Sci-
entific Publishers Limited; 2001.
3. Heneghan C, Badenoch D. Evidence-based Medicine Toolkit.
Oxford, UK: Blackwell Publishing Limited; 2006.
4. Mayer D. Essential Evidence-Based Medicine. Cambridge,
UK: Cambridge University Press; 2004.
5. U.S. Preventive Services Task Force. Guide to Clinical Pre-
ventive Services. 2nd ed. Baltimore, MD: Williams &
Wilkins; 1996.
6. The Canadian Task Force on the Periodic Health Exami-
nation. The Canadian Guide to Clinical Preventive Health
Care. Ottawa: Health Canada; 1994.
7. Cook DJ, Guyatt GH, Laupacis A, et al. Rules of evidence
and clinical recommendations on the use of anti-
thrombotic agents. Chest. 1992; 102(4 suppl):305S–311S.
8. Sackett DL. Rules of evidence and clinical recommenda-
tions. Can J Cardiol. 1993; 9:487–489.
9. Guyatt G, Rennie D, eds. User’s Guides to the Medical Lit-
erature. Chicago, IL: AMA Press; 2002.
10. Cook DJ, Guyatt GH, Laupacis A, et al. Rules of evidence
and clinical recommendations on the use of anti-
thrombotic agents. Chest. 1992; 102(4 suppl):305S–311S.
Chapter 7
Develop a
Conclusion and
Recommendation
Patrick J. Bryant
Step 5 – Develop a Conclusion and Recommendation
Organizing the Pertinent Information
Developing a Conclusion from the Summary
Developing a Recommendation from the Conclusion
From Conclusion to Recommendation
Type of Decision
Confidence of the Recommendation
Format for the Recommendation
Support for the Recommendation
Summary
References
Step 5 – Develop a Conclusion and
Recommendation
The fifth step of the 5-Step Evidence-Based Medicine Process, De-
velop a Conclusion and Recommendation, may be the most impor-
tant task and yet can be the most challenging for the practitioner.
This is the step that provides a solid and defendable recommenda-
tion. The degree of confidence in the recommendation and the jus-
tifications supporting that recommendation are based on the quality
of evidence available. Essentially, the recommendation is as strong as
the supporting evidence. A recommendation developed following
the 5-Step Evidence-Based Medicine Process is easy to support and
defend because the highest quality of evidence has been utilized.
103
104 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
A recommendation developed following the
5-Step Evidence-Based Medicine Process is
Key easy to support and defend when necessary.
Idea Essentially, the recommendation is as strong
as the supporting evidence.
Organizing the Pertinent Information
Once the evidence has been retrieved, analyzed, and categorized by
quality, sorting the pertinent information by level of evidence is ex-
tremely helpful to begin developing a conclusion. By organizing the
evidence from the highest to lowest in confidence (levels of evidence
and major limitations), the practitioner can begin to identify the
most reliable information to develop a conclusion (see Figur
Figuree 7.1
7.1).
Level I Level I Level II Level III Level IV Level V
minor major major major major major
limitations limitations limitations limitations limitations limitations
Highest Lowest
Confidence Confidence
Figur
iguree 7.1: Sort Pertinent Studies by Level of Evidence and Major Limita-
tions
Organizing pertinent information by level of
Key evidence and using a tool called Summary
Idea Table of Evidence Evaluated brings clarity
to what the evidence is supporting.
The studies with the highest confidence can be organized even
further using the Summary Table of Evidence Evaluated (see table
in Figur
Figuree 7.2 and also the “EBM Tool Kit” at the end of this
book). This table summarizes not only the level of evidence, but
also the study outcome and major limitations from the Ten Ma-
jor Considerations identified in Step 3: Evaluate Literature. Clar-
Chapter 77: Develop a Conclusion and Recommendation 105
Clinical Question - Is Drug A as efficacious and safe as Drug
B, currently on formulary?
Level of Major
Article Outcome
Evidence Limitations
Study #1 Equivalent I No Major
Study #2 Equivalent II Major-power
Study #3 Equivalent II Major-power
Figur
iguree 7.2: Summary Table of Evidence Evaluated
ity emerges by observing all the evidence presented in the same
table at the same time.
Developing a Conclusion from the Summary
The conclusion becomes more apparent as results from the high-
est level of evidence are compared and contrasted in the Sum-
mary Table of Evidence Evaluated. By reviewing these findings in
more detail, the practitioner will be able to identify the differ-
ences between the highest quality trials. These differences are
important in fine-tuning the conclusion and one’s degree of con-
fidence in that conclusion.
Example 7.1. Developing a Conclusion
Clinical Question - Is Drug A as efficacious and
safe as Drug B, currently on formulary?
Summary Table of Evidence Evaluated
Article Outcome Level of Major
Evidence Limitations
Study #1 Equivalent I No Major
Study #2 Equivalent II Major-power
Study #3 Equivalent II Major-power
and dose for
Drug A
106 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
For example, a conclusion that Drug A has equiva-
lent efficacy compared to Drug B can be made from
the single Level I trial with no major limitations.
The major limitation of power not being met puts
the first Level II trial in question. When power
has not been met and there is no difference be-
tween treatment groups, the concern is that a dif-
ference may have been shown if enough patients
had been entered to meet power (see Chapter 2:
Basics for Interpretation).
After careful review, it is determined that there
is no way to assess whether this major limitation
had an effect on the overall study results; however,
one should assume that it did affect the overall re-
sults. The second Level II trial has this same major
limitation in addition to a question about whether
the higher dose used for Drug A may have given
that drug an unfair advantage. If a normal therapeu-
tic dose of Drug A would have been used, Drug A
may have been less effective than Drug B. There is
no way to determine if this major limitation had an
effect on the overall study results. To be on the con-
servative side, one should assume that this major
limitation did have an effect on the overall results.
This example illustrates the necessity for including the major
limitations to help determine overall confidence in a study’s results.
If major limitations are not taken into consideration, an inappropri-
ate conclusion may be reached. Even with a high level of evidence,
overall reliability of the trial could be reduced by one major limita-
tion such as dissimilar groups after randomization. (For a list of the
Ten Major Considerations, see Chapter 5: Evaluate Literature.)
Key Incorporating the identified major limitations assists
Idea in determining overall reliability of the evidence.
Chapter 77: Develop a Conclusion and Recommendation 107
It is crucial to determine whether the results appear to be
affected by an identified major limitation. After this determina-
tion, a decision can be made to either include the trial results as
reliable or discredit findings and exclude the trial. For instance,
in Example 7.1, if Study #1 had dissimilarity in demographics
between treatment groups (e.g., height of the patients), this has
nothing to do with efficacy outcomes in this case. It would be
appropriate to conclude that Drug A and Drug B are equivalent
regarding efficacy. This case illustrates the importance of deter-
mining the impact each identified major limitation could have
on overall study results.
Developing a Recommendation from the
Conclusion
From Conclusion to Recommendation
Even the simplest decision involves a process that must occur
between forming a conclusion and developing a recommenda-
tion. Three components are involved in this process (see Figur
Figuree
7.3
7.3).
Quality of the Logical Clinical
Evidence* + Reasoning + Judgment =
Formation of a Recommendation
*Quality of the evidence, major strengths, and any major limitations are considerations.
Figur
iguree 7.3: Components Required to Develop a Recommendation
The first component (quality of the evidence) is a result of
Step 3: Evaluate Literature and Step 4: Categorize the Quality of
Evidence of the 5-Step Evidence-Based Medicine Process out-
lined in this book. The quality of the available evidence is con-
sidered in addition to major strengths and limitations of studies
that make up this available evidence. When developing a recom-
108 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
mendation, the practitioner should know that the overall reli-
ability of the evidence is important.
Logical reasoning, the second component, requires both de-
ductive and inductive reasoning. This component provides prac-
ticality in forming the recommendation. For instance, when no
head-to-head comparative trials exist for two drugs, other trials
with identical control groups for each drug in similar patient
populations may be used along with logical reasoning to draw
conclusions that go into the recommendation.
The third component, clinical judgment, comes from expe-
rience as a clinical practitioner. Clinical judgment fills in gaps
where evidence does not exist or is weak. For instance, a
practitioner’s clinical experience reveals that with a specific pa-
tient, drugs from the same pharmacological class have different
degrees of efficacy. Based on this clinical judgment, the practitioner
may suggest changing to a different drug in the same pharmaco-
logical class to assist in identifying the optimal agent.
Conclusions drawn from the evidence analyzed assist
Key in the development of recommendation statements.
Reliability of the evidence combined with logical
Idea
reasoning and clinical judgment all factor into the
clinical decision making process.
For extremely complex decisions leading to recommenda-
tions, formal decision analysis processes are used.1–4 The pri-
mary tool utilized is the decision tree. Discussion on the use of
decision trees and formal decision analysis processes are beyond
the scope of this book. A detailed tutorial on performing this
type of analysis is available.5
Type of Decision
Two types of clinical decisions will be discussed in this book—indi-
vidual patient and population-based. An individual patient deci-
Chapter 77: Develop a Conclusion and Recommendation 109
sion is represented by a typical treatment plan developed for a single
patient. A population-based decision is represented by a formulary
management clinical decision (see table in Figur
Figuree 7.4 , page 111).
7.4
Both decisions are important and will affect health care for patients.
The population-based decision is typically made in a more
conservative manner since many patients will be affected and little
is known about the specifics of each patient in this population.
When details about a single patient’s disease and medication his-
tory are known, the practitioner can customize the decision to fit
that individual patient. Knowing these specifics allows the practi-
tioner to use clinical judgment associated with that individual. With
an understanding of the type of clinical decision being made (indi-
vidual patient versus population-based), the practitioner can modify
the final recommendation to fit the type of decision.
Population-based decisions require a more
Key conservative approach since specifics
Idea about the individual patients that will be
affected are unknown.
Example 7.2. Developing a
Recommendation Based on T ype of
Type
Decision
Clinical Question – Is Drug A as efficacious and
safe as Drug B, currently on the formulary?
Her
Heree is what we know fr om the evidence:
from
1. Conclusion from evidence – Drug A and Drug
B have equivalent efficacy and safety with no
specific advantage of one over the other. Drug
A is considerably more expensive than Drug B.
2. This is supported by a Level I trial with no
major limitations.
3. The two Level II trials are not considered since
110 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
they were not powered to show a difference,
if one existed and no difference between treat-
ment groups was noted.
4. This is a population-based decision; therefore,
a conservative stance needs to be taken with
the clinical decision to be made here. If this
were an individual patient decision, the prac-
titioner would want to assess any subgroup
analyses that may pertain to the specific pa-
tient. With this in mind, a completely differ-
ent recommendation could be required.
Recommendation Statement: I do not recommend
adding Drug A to the formulary. (Note: The degree
of confidence one has in this recommnedation still
needs to be fully determined to adjust the overall
tone of the statement. This will be explained in the
next section.)
Confidence of the Recommendation
The confidence of a recommendation depends on the quality of
evidence supporting that recommendation statement. The degree
of confidence one has in the evidence is important when deter-
mining how strongly to state the recommendation. To assist in
determining strength of the recommendation statement, the Grade
of Recommendation Tool is used. The highest level of evidence
(Level I) is also the evidence that carries the greatest confidence
(see Figur
Figuree 7.5, page 112, and also the “EBM Tool Kit” at the end
7.5
of this book). For instance, a Grade A Recommendation would be
based on Level I evidence. Unless major limitations exist that would
cause concern with the study’s integrity, this grade allows a strong
and firm recommendation statement.
The Grade of Recommendation Tool helps
Key
establish the firmness of the language used
Idea in the recommendation statement.
Chapter 77: Develop a Conclusion and Recommendation 111
Type of
Clinical Decision Manner in Which Decision Is Made
Less Conservative More Conservative
Individual • Only one patient in-
Patient volved.
Decision • The patient’s medical
history and back-
ground are known in
addition to drug aller-
gies.
• Specific co-morbid dis-
ease states such as liver
and/or kidney impair-
ment are known.
• Prognosis is known for
this specific patient.
• Also known is whether
the drug in question is
the last option to treat
the disease for this pa-
tient.
Population- • Potentially, a large num-
Based ber of patients will be
Decision affected.
• Individual patient medi-
cal history and back-
ground are not known,
including drug allergies.
• Specific co-morbid dis-
ease states are un-
known.
• Prognosis of each pa-
tient in the population
is not known.
• Also not known is
whether the drug in
question is the last
treatment option for
each patient.
Figur
iguree 7.4: Individual Patient versus Population-Based Decisions
112 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Level of Evidence (LOE) Grade of Recommendation
GRADE A
LEVEL I
Level I
confidence
Level II GRADE B
LEVEL II
Level III
GRADE C
Level IV LEVELS III,
IV & V
Level V
Figur
iguree 7.5: Correlation among Confidence in Evidence, Level of Evidence,
and Grade of Recommendation
Example 7.3. Str ength of
Strength
Recommendation Statement Based on
Confidence in Evidence
Level of Evidence Grade of Recommendation
GRADE A Study #1
LEVEL I
Level I Study #2
confidence
Level II GRADE B Study #3
LEVEL II
Level III
GRADE C
Level IV LEVELS III,
IV & V
Level V
Taking the previous example where the conclu-
sion was that Drug A and Drug B have equivalent
efficacy based on a Level 1 study with no major
limitations, it is clear that this Level of Evidence
provides a Grade A Recommendation. This is the
highest Level of Evidence available to support the
Chapter 77: Develop a Conclusion and Recommendation 113
recommendation and, therefore, a strong and firm
tone should be used in the recommendation state-
ment if no major limitations exist.
Knowing that a Grade A Recommendation desig-
nation allows the highest degree of firmness in
the language used to develop the recommenda-
tion statement, a firm recommendation statement
can now be developed for this situation. For in-
stance, I strongly recommend to not add Drug A
to the formulary.
Format for the Recommendation
Recommendations should include a statement with justification
Figuree 7.6 and
points that support the recommendation (see Figur
also the “EBM Tool Kit” at the end of this book). Although vari-
ous formats can be used to present a recommendation, a clear
and concise statement is important to ensure effective communi-
cation. This template can be modified to fit various styles, but
two primary components should be present—a clear and con-
cise recommendation statement followed by supportive justifi-
cation points.
THE RECOMMENDATION STATEMENT
JUSTIFICATION IN A BULLET FORMAT
• Efficacy
• Safety
• Other special considerations/populations
• Cost
Figur
iguree 7.6: Format for a Recommendation
114 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Example 7.4. The R ecommendation
Recommendation
Statement
I strongly recommend that Drug A not replace
Drug B on the formulary, but be reserved as sec-
ond-line therapy for allergic conjunctivitis only
after Drug B has been tried and failed.
Support for the Recommendation
Justification points provide clear and concise support, similar to
“pillars,” for the recommendation statement (see Figur Figuree 7.7
7.7). Each
justification point includes the efficacy, safety, special considerations/
special populations, and cost associated with a specific situation.
Information obtained from the evidence should be addressed un-
der each of these considerations:
Efficacy
Summary of the highest level of evidence trial(s) being
used to develop the recommendation statement.
Specifically what that evidence confirms. Any major limi-
tations identified along with the determined impact that
those limitations may have on the results, especially the
primary outcome measure(s).
Any outcomes that are of interest to the clinical question.
Safety
Description of adverse drug reactions noted especially
those different from the comparison group(s).
A statement of overall safety profile and any safety
concern(s). This should be obtained from information
that includes the greatest number of patients exposed
(usually, the Package Insert/Prescribing Information) to
attempt to identify rare adverse drug reactions.
Special considerations/special populations
This is a very broad scope category, and the content is
Chapter 77: Develop a Conclusion and Recommendation 115
RECOMMENDATION
Other
Efficacy Safety Consid- Cost
erations
Figur
iguree 7.7: Justification Points Support the Recommendation
highly dependent upon the defined clinical question.
Some types of special considerations that would be ap-
propriate to include are differences in dosing convenience
and route of administration between the drugs in ques-
tion.
Any knowledge gained that addresses the advantages and/
or disadvantages of the study drug over existing therapy.
Specific populations that need to be highlighted due to
the drug’s benefit or disadvantages on those populations
should be addressed.
Any knowledge gained from the study that provides an
understanding of how and where this drug fits in overall
therapeutic strategies and standard practice guidelines
should be included.
Cost
Differences in cost between the treatments studied includ-
ing nursing/pharmacy and other related costs, if possible.
Differences in cost compared to current standard of care
treatment.
Any impact associated with these differences in cost (both
savings and/or expenditures).
116 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
The following example illustrates effective use of justification
points to support/defend the previously developed recommen-
dation statement.
Example 7.5. Justification for the
Recommendation
I recommend that Drug A not replace Drug B on
the formulary, but be reserved as second-line
therapy for allergic conjunctivitis only after Drug
B has been tried and failed. The following reasons
support this recommendation:
Ef ficacy
Efficacy One Level I trial with no major limitations
confirms that Drug A and Drug B are
equivalent in efficacy.
Two additional Level II trials supported the
above finding. Major limitations that make
these results less reliable include 1) no
power calculation for either trial and 2) in
one trial higher than FDA-recommended
doses of Drug B were used.
Safety Safety results from this trial are similar to
the Prescribing Information available for
each drug and confirms that the safety pro-
files are similar for both drugs with the ex-
ception that Drug A has a higher incidence
of burning and stinging of the eyes.
Special No special considerations have been iden-
Consider--
Consider tified. There are no special population is-
ations/ sues identified.
Special
Populations
Cost Cost for Drug A is considerably higher than
for Drug B since Drug B has a generic
equivalent.
Chapter 77: Develop a Conclusion and Recommendation 117
Note: Level of Evidence is a way to communicate with other
individuals who know this 5-Step Evidence-Based Medicine Pro-
cess; however, be prepared to describe each level in a manner
that others who are not familiar with this method to categorize
quality can understand. For instance, in Example 7.5, the effi-
cacy justification section could be rewritten in more generic terms
such as, “One high quality, randomized controlled trial meeting
power with no major limitations confirms that Drug A and Drug
B are equivalent in efficacy.”
Summary
Developing a conclusion and recommendation can be challeng-
ing, but the result is a reproducible and defendable recommen-
dation when it is done based on evidence. Utilizing the Sum-
mary Table of Evidence Evaluated tool, the pertinent informa-
tion can be organized in a manner so that a clear conclusion be-
comes apparent. Knowing the type of decision being made will
help to determine if the recommendation must remain conserva-
tive. A more conservative recommendation should be developed
for population-based decisions since there are considerably fewer
specifics known in this situation than with an individual patient
decision. The level of evidence and presence of major limitations
will assist in determining the confidence one can place in the
recommendation.
Three separate components are involved when developing a
recommendation statement: quality of the evidence, logical rea-
soning, and clinical judgment. The recommendation statement
should be supported by justification points addressing efficacy,
safety, special considerations/special populations, and cost. Es-
sentially, the recommendation is as strong as the supporting evi-
dence. Using the 5-Step Evidence-Based Medicine Process de-
scribed in this book provides a recommendation that is easy to
support and defend.
118 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
References
1. Gross R, ed. Decisions and Evidence in Medical Practice:
Applying Evidence-Based Medicine to Clinical Decision Mak-
ing. St. Louis, MO: Mosby; 2001.
2. Mayer D, ed. Essential Evidence-Based Medicine. New York,
NY: Cambridge University Press; 2004.
3. Jenicek M. Foundations of Evidence-Based Medicine. New
York, NY: The Parthenon Publishing Group Inc.; 2003.
4. Sarnes M. Making a Pharmacotherapy Decision Using a
Decision–Analytic Framework. In: Chiquette E, Posey LM,
eds. Evidence-Based Pharmacotherapy. Washington, D.C.:
American Pharmacists Association; 2007.
5. Detsky AS, Naglie G, Krahn MD, et.al. Primer on medical
decision analysis: Part 1—getting started. Med Decision
Making. 1997; 17(2):123–125.
Chapter 8
Alternative Sources
of Evidence
Heather A. Pace
Alternative Sources of Evidence
Systematic Reviews and Meta-Analyses
Narrative Reviews
Practice Guidelines
Summary
References
Alternative Sources of Evidence
This chapter presents alternative sources of evidence used for
clinical decision making and outlines the strengths and weak-
nesses of each. Systematic reviews, meta-analyses, narrative re-
views, and practice guidelines are useful tools, but without ap-
plying the process of evidence-based medicine they can lead to
inappropriate and possibly dangerous clinical decisions.
Systematic Reviews and Meta-Analyses
Systematic reviews and meta-analyses are methods used to sum-
marize large numbers of clinical trials and, if used carefully, can
be valuable tools for the busy practitioner. The general process
for systematic reviews and meta-analyses is similar. Both meth-
ods evaluate and combine results from a compilation of random-
ized controlled trials pertaining to the specific clinical question
at hand. The key difference between these two methods is in the
outcome and how each reaches its conclusion. Meta-analyses
employ statistical methods to combine results of primary trials
and produce a single pooled estimate of an intervention’s effect.
119
120 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Meta-analyses establish criteria to ensure that the data from single
trials are homogenous to allow for the creation of new data from
statistical analysis, whereas systematic reviews do not produce
new data, merely a combination of the existing data from the
single trials. It is important to consider the quality of the new
data produced by a meta-analysis as this can affect the overall
reliability of the meta-analysis results. Systematic reviews do not
employ statistical methods and produce qualitative results rather
than quantitative results. Both methods can produce strong evi-
dence or potentially poor quality evidence; therefore, caution is
advised and some form of critical evaluation is required to deter-
mine the quality of this evidence.
Systematic reviews and meta-analyses can be useful in the
following cases:
When primary data is conflicting or in disagreement as
to the direction or magnitude of effect
If sample sizes in primary research are too small to detect
an effect
If large trials simply are not feasible
Example 8.1. Using Multiple Studies
“Does estrogen replacement therapy increase the
risk of breast cancer?” Because the incidence of
breast cancer is so low, it may not occur often
enough to answer the question based on individual
studies. Combining multiple studies may provide
sufficient incidence to answer the question.
Oftentimes meta-analyses can provide conclusions about a
treatment effect that cannot be drawn from individual trials and
may possibly answer questions not addressed in single trials. The
purpose of conducting a meta-analysis is to:
Increase statistical power, resolve uncertainty when single
trials disagree
Chapter 88: Alternative Sources of Evidence 121
Improve estimates of size effect
Answer new questions (use the old data in a new way)
Bring about improvements in quality of research
Meta-analyses employ statistical methods to
combine results of primary trials and produce a
Key single pooled quantitative estimate of an
Idea intervention’s effect (new data produced),while
systematic reviews produce non-statistical,
qualitative results (no new data produced).
Systematic reviews and meta-analyses can be described as
scientific investigations with predefined methods and original
studies as the subjects.1 As mentioned earlier, quality needs to be
analyzed just as with single trials. The methods to determine the
quality of systematic reviews and meta-analyses are similar to
those used for individual clinical trials and are described in Chap-
ter 5: Step 3–Evaluate Literature. Similar considerations as those
used for clinical trials are examined to determine the quality of
the resulting evidence provided by systematic reviews and meta-
analyses. Certain design characteristics that should be evaluated
include the following2:
Is there a clear objective stated?
Was a comprehensive search used to identify all relevant
studies? (recruitment methods)
Are the inclusion/exclusion criteria for the selection of
studies appropriate?
Are the individual studies evaluated for validity and quality?
Are the individual study characteristics described?
Is there an appropriate quantitative data synthesis? (ap-
propriate statistical analysis)
Are both clinical and statistical significance reported?
Are the results generalizable to clinical practice?
122 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
While systematic reviews and meta-analyses can offer advan-
tages over single studies, they are not as useful for patient-spe-
cific questions. Because of the potential in design flaws, intro-
duction of bias, or overall false conclusions, results from system-
atic reviews and meta-analyses should be applied with caution
and should not be the sole source of evidence when making a
clinical decision.
Narrative Reviews
Narrative or non-systematic reviews lack an orderly method for
identifying and analyzing data. The biggest difference between
narrative reviews and systematic reviews or meta-analysis is that
narrative reviews do not provide pooled results from primary
trials. Narrative reviews often provide a broad overview of a spe-
cific topic, including disease pathology, diagnosis, prevention,
and multiple therapeutic interventions. Conclusions can be less
evidence-based and affected by the opinions of the authors.
Oftentimes considered a tertiary source of literature, narrative
reviews are useful in establishing background information but
should not be used as supporting evidence for a recommenda-
tion. Moreover, narrative reviews often contain extensive bibli-
ographies and can provide an excellent source for identifying lit-
erature surrounding a specific subject. The table in Figur
Figuree 8.1
provides an overview of systematic reviews, meta-analysis, and
narrative reviews.
Practice Guidelines
A lack of understanding about the requirements for
guideline development could lead to either
Key inappropriate interpretation or acceptance of
Idea inappropriate enforcement levels of biased
guideline recommendations.2
Chapter 88: Alternative Sources of Evidence 123
Narrative Systematic Meta Analysis
Featur
Featuree Review Review
Clinical Often broadly Clearly defined Clearly defined and
question defined and focused focused
Literature search Methods not Predefined Predefined strategy,
usually explicitly strategy, explicit explicit and
described and comprehensive comprehensive
Studies included Inclusion Predefined Predefined inclusion
methods not inclusion and and exclusion criteria
usually described exclusion criteria
Unpublished Not usually Possibly Possibly
literature included
Blinding of No Yes Yes
reviewers
Analysis of data Variable and Rigorous and Rigorous and objective—
subjective no new new data pr
objective—no oduced
produced
data pr oduced
produced
Results statistically No No Yes
evaluated
Type of results Qualitative Qualitative Quantitative
Adapted from Pinsky L, Deyo R. Clinical guidelines: a strategy for translating evidence into
practice. In: Geyman J, Deyo R, Ramsey S, eds. Evidence Based Clinical Practice. 1st ed.
Woburn, MA: Butterworth-Heinemann; 2000:119–123.
Figur
iguree 8.1: Comparisons of Characteristics of Reviews
Practice guidelines help to provide consistent health care, estab-
lish practice standards, and aid in guiding clinical decisions. Prac-
tice guidelines can be flawed, making it important to evaluate
and apply certain evidence-based medicine principles. The in-
formation used to support and develop the guidelines should be
evaluated as well as the overall quality of the guideline.
The first consideration when evaluating a guideline is to de-
termine the method used to develop it. Not all practice guide-
lines are evidence-based; however, many guidelines are moving
towards an evidence-based process. The reasons for this shift
124 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
include a rapid increase of available information, greater avail-
ability of treatments, an increase in the number of technologies
of unproven efficacy, and wide variations in practice habits among
physicians.3
Guidelines can be evidence- or consensus-based. Evidence-
based guidelines follow a rigorous process to identify and evalu-
ate literature for developing recommendations, whereas a con-
sensus process relies on the experience of experts to develop and
support recommendations. ATP III (Adult Treatment Panel III)
and JNC7 (the Seventh Report of the Joint National Committee
on Prevention, Detection, Evaluation, and Treatment of High
Blood Pressure) are examples of some of the most widely used
evidence-based guidelines.
Although an evidence-based development process is preferred,
consensus can be useful in situations where limited evidence or
information exists. In these cases, guidelines can help establish
practice standards. Guidelines can be a hybrid of the two meth-
ods, combining both evidence-based and expert opinions. This
approach can be useful when practice trends are needed to fill-in
“gaps” left by poor evidence or lack of evidence. The table in
Figur
Figuree 8.2 represents the differences between the two types of
guidelines.
Evidence-Based Guidelines Consensus Guidelines
Principles of evidence-based medicine Principles of clinical observation
Reasoning based on clinical studies Reasoning based on pathophysiology
Guidelines based on evaluation Guidelines based on expert opinion
of medical literature
Figur
iguree 8.2: Differences Between Evidence-Based and Consensus
Guidelines
Credentials of the authors and the source of the document
are important areas to consider when evaluating the quality of
practice guidelines. Ideally authors or an organization are ex-
perts in the specific field. For instance, the American Diabetes
Association and the American College of Obstetricians and Gy-
Chapter 88: Alternative Sources of Evidence 125
necologists are considered examples of reputable organizations
and should be expected to produce reliable, accurate guide-
lines; however, one should still determine the method used to
develop the guideline: evidence-based, consensus-based, or
hybrid.
Practice guidelines should also be periodically updated to
keep up with changing practice, new treatments, and recent
evidence. The incorporation of new data in updates strength-
ens the recommendation and value of the guideline. It is im-
portant to verify the date of development of a guideline to en-
sure recommendations are based on the most recent evidence
and treatments available.
When evaluating practice guidelines, consider the following
questions:
Development method of guideline (consensus versus evi-
dence-based)?
Are the recommendations valid and reliable?
Are the recommendations generalizable?
Was there a peer review process?
Were literature search methods outlined and explained?
Are the guidelines reviewed and updated on a regular
basis?
Summary
The evidence-based medicine process is not only useful and im-
portant in evaluating single randomized controlled trials, but it
is also helpful to determine the quality and reliability of alterna-
tive sources of evidence used for clinical decision making. The
application of this process will ensure that information is used
appropriately to support clinical decisions and recommendations.
Without the use of this process, systematic reviews, meta-analy-
ses, narrative reviews, and practice guidelines could potentially
lead to inappropriate and dangerous clinical decisions.
126 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
References
1. Cook DJ, Mulrow CD, Haynes RB. Systematic reviews:
synthesis of best evidence for clinical decisions. Ann In-
tern Med. 1997; 126:376–380.
2. Moores K. Evidence-Based Clinical Practice Guidelines.
In: Malone P, Kier K, Stanovich J. Drug Information: A Guide
for Pharmacists. 3rd ed. McGraw Hill; 2006:289–338.
3. Lewis TA, Fineberg HV, Mosteller F. Findings for public
health from meta-analysis. Ann Review of Public Health.
May 1985; 6:1–20.
Chapter 9
Applications
Lindsey N. Schnabel & Heather A. Pace
Applications
Incorporating Evidence-Based Clinical Decision
Making into Clinical Practice
Individual patient decisions
Population-based decisions
Examples
Summary
Applications
Incorporating Evidence-Based Clinical Decision
Making into Clinical Practice
How do practitioners incorporate evidence-based decision mak-
ing into clinical practice? By following the five-step process out-
lined throughout this book, pharmacy practitioners can assess
the evidence available and make either 1) firm recommendations
and decisions based on results of rigorously controlled investiga-
tions or 2) cautious recommendations and decisions when re-
sults of uncontrolled clinical observations exist (Figur
(Figuree 9 .1)
9.1)
.1).
Evidence-based medicine, when used in pharmacy, is applied in
various situations. This chapter will examine several examples of
how pharmacy practitioners can use evidence-based medicine in
clinical practice when making both individual patient decisions
as well as population-based decisions.
127
128 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Make
Make FIRM CAUTIOUS
recommendations recommendations
& decisions based & decisions when
on results of only results of
rigorously uncontrolled clinical
controlled observations
investigations exist
iguree 9.1: Explanation of Differences in Firm and Cautious Recommenda-
Figur
tions Based on Trial Type
Individual patient decisions
Individual patient recommendations differ from
Key
population-based decisions in that individual patient
Idea decisions require a less conservative approach.
Individual patient decisions are made frequently in pharmacy
and often require a less conservative approach due to the fact that
more information is available regarding an individual versus an
entire population. Practitioners must look at the patient as a whole
and consider many factors when making a recommendation or
decision regarding an individual’s health. As discussed previously,
it is imperative to consider efficacy, safety, special considerations/
special populations, and cost when making a clinical decision or
recommendation. This also applies to individual patients.
The clinician should apply the 5-Step Evidence-Based Medi-
cine Process to determine if the treatment option is efficacious.
Various sources of evidence should be reviewed such as
interventional or observational clinical trials, case control stud-
ies, case series, or individual case studies. By assessing risk ver-
sus benefit, the practitioner can determine if the efficacy of the
therapy outweighs the harm if any it may cause to the patient.
Chapter 99: Applications 129
Other important aspects to consider when looking at the safety
justification of a patient-specific decision are allergies, concomi-
tant medications and potential drug interactions, current disease
states and interactions that may be associated with them, and
compliance/adherence concerns.
Special populations that should be considered include dos-
ing convenience, route of administration, specific situations that
may affect the patient positively or negatively, and advantages or
disadvantages over lack of treatment or existing treatment. Spe-
cifically, “Is there a population that may especially benefit from
the recommendation?” An angiotensin-converting enzyme inhibi-
tor, for instance, would benefit a kidney failure patient who also
has hypertension.
Finally, cost should be considered. “Can the patient afford
the treatment?” “Are there patient assistance programs available
to help with affordability of the therapy?” “Is there a significant
cost associated with the treatment such as travel expenses or
medical devices used for administration?” “Would any additional
cost be outweighed by preventing future complications or mor-
bidity or mortality?”
By factoring efficacy, safety, special considerations/special
populations, and cost into forming a decision as well as assessing
risk versus benefit, practitioners can be assured that an appro-
priate individual patient decision is made. Chapter 7: Step 5 –
Develop a Conclusion and Recommendation provides additional
factors to consider for each of these catagories.
Population-based decisions
Key Population-based recommendations differ from
individual patient recommendations in that population
Idea
decisions require a more conservative approach.
Population-based clinical decisions differ from individual ones
in that a more conservative approach is required because fewer
130 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
specifics are known. When formulating a recommendation for a
population such as a medication class review, comparative analy-
ses, or a formulary request, the clinician’s decision is affecting a
large number of people who have many different disease states,
different medication regimens, and different compliance issues.
These decisions, similar to individual patient decisions, also re-
quire the practitioner to consider efficacy, safety, special consid-
erations/special populations, and cost when making a recommen-
dation.
The efficacy justification to support population-based deci-
sions often is derived from evidence such as interventional or
observational clinical trials, case control studies, or well designed
meta-analyses or systematic reviews. The safety considerations
of a population-based decision should be assessed by reviewing
the evidence as well as the most current prescribing information.
As with individual clinical decisions, it is imperative that the
following special considerations/special populations are taken into
account for population-based recommendations: dosing conve-
nience, route of administration, specific situations that may af-
fect the population positively or negatively, and advantages or
disadvantages of the treatment.
When formulating a recommendation for a population, prac-
titioners have fewer known specifics regarding prescription cov-
erage, patient income, and other variables; therefore, cost is typi-
cally one of the final components to consider. Sometimes the
question is asked, “How much better should one drug be in terms
of efficacy, safety, or special considerations/special populations to
justify the additional expense?” If efficacy, safety, and special con-
siderations/special populations are the same between compara-
tive drugs, one can consider cost to be the primary determinant.
Chapter 7: Step 5 – Develop a Conclusion and Recommendation
provides additional factors to consider for each of these catagories.
Examples
The following examples are provided to illustrate application of
the evidence-based process described in this book to both indi-
vidual patient and population-based decisions.
Chapter 99: Applications 131
Example 9.1. Individual Patient
Patient
Clinical Decision/R ecommendation
Decision/Recommendation
This example illustrates how to use the 5-Step
Evidence-Based Medicine Process to assist in mak-
ing a clinical decision/recommendation for an in-
dividual patient.
A physician wants to give BP, a male patient, Drug
A for treatment of his osteoporosis instead of Drug
B because Drug A can be given once per month
whereas Drug B requires weekly administration.
Somewhere the pharmacist read or heard that
Drug A was not as effective in male osteoporosis
as Drug B. Before the pharmacist calls the physi-
cian to discuss this issue, he or she should con-
duct an evidence-based medicine analysis to con-
firm that Drug A is not as effective as Drug B in
male osteoporosis.
Patient Backgr ound:
Background:
63-year-old male
No known drug allergies
Past medical history:
Hypertension—controlled on lisinopril
Type 2 diabetes—controlled on metformin and
insulin glargine
Hypercholesterolemia—uncontrolled on
simvastatin; ezetimibe added at most recent
visit with physician
Prescription coverage that discounts all medica-
tions
132 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Step 1: Define the Clinical Question
Determine the actual question being asked so
that an appropriate response is formed. For
this example, the question would be “Is Drug
A as effective as Drug B in the treatment of
male osteoporosis for BP?”
Step 2: Retrieve Pertinent Information
A literature search was performed using two da-
tabases and the following search terms: Drug A
(brand and generic names), Drug B (brand and
generic names), osteoporosis, male. After review-
ing the search results from both databases using
appropriate limits and the search and sift tech-
nique, the clinician found two key articles. The
clinician performed a bibliographic search by re-
viewing the references of the two articles found.
This technique led to a case series. Overall, the
literature search revealed the following articles:
Article #1 – A randomized, double-blind, par-
allel group study in male patients with os-
teoporosis compared Drugs A and B. Power
was set and met; that is, sample size was ad-
equate to show a difference between study
groups, if a difference really exists. One major
limitation was noted; the groups were not simi-
lar after randomization. No other limitations
were identified from the ten major consider-
ations list. The results of the study showed
that Drug B was superior to Drug A for the
treatment of male osteoporosis, and this dif-
ference was statistically significant (p=0.001).
There was no difference in the incidence or
severity of adverse drug reactions between the
Chapter 99: Applications 133
two drugs. No special considerations/special
populations were noted.
Article #2 – A randomized, double-blind,
crossover study in male patients with os-
teoporosis compared Drugs A and B for treat-
ment. Power was set and met, thus indicating
an adequate sample size has been used to show
a difference between Drugs A and B, if a dif-
ference really exists. No other major limita-
tions were noted. The results of this study
showed that Drugs A and B are equivalent in
efficacy for the treatment of male osteoporo-
sis (p=0.26). In addition, no difference in the
incidence or severity of adverse drug reactions
between the two drugs was noted. No special
considerations/special populations were iden-
tified. The two drugs were similar in price.
Article #3 – A case series of male patients with
osteoporosis were studied. These patients were
observed over a 1-year period after being placed
on Drug A by the primary physician. There was
an improvement noted in bone density tests
after 1 year of receiving Drug A. Some adverse
drug reactions were noted, but they were an-
ticipated based on the package insert informa-
tion and were not severe. No special consider-
ations/special populations were noted.
The two drugs are similar in price; however,
Drug A is covered by prescription insurance.
Step 3: Evaluate Literatur
Literaturee
Efficacy
• Critically evaluate each trial and determine
if the article supports or refutes the use of
Drug A over Drug B.
134 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
-
Article #1 – The results of the study
showed that Drug B was superior to
Drug A for the treatment of male os-
teoporosis, and this difference was sta-
tistically significant (p=0.001).
- Article #2 – The results of this study
showed that Drugs A and B are equiva-
lent in efficacy for the treatment of
male osteoporosis (p=0.26).
- Article #3 – There was an improvement
noted in bone density tests after 1 year
of receiving Drug A; however, statis-
tics were not performed since there
was no comparison group.
• Establish if each trial has major or minor
limitations.
- Article #1 – The groups were not simi-
lar in baseline bone mineral density
scores after randomization, and this
appeared to have potentially affected
the results. No other major limitations
were identified.
- Article #2 – No major limitations from
the ten major considerations list were
noted.
- Article #3 – This study was a case se-
ries and, therefore, did not have a com-
parative group. Due to this study de-
sign, several of the ten major consid-
erations were considered to be major
limitations.
Safety
• Assess risk versus benefit of Drug A ver-
sus Drug B versus no treatment.
Chapter 99: Applications 135
-
Article #1 – No difference was noted
in the incidence or severity of adverse
drug reactions between the two drugs.
- Article #2 – No difference was noted
in the incidence or severity of adverse
drug reactions between the two drugs.
- Article #3 – Some adverse drug reac-
tions were noted, but they were antici-
pated based on the package insert in-
formation for Drug A and were not se-
vere. Note there was no comparison
group.
Special considerations/special populations
• Establish if Drug A is dosed more or less
conveniently than Drug B.
- Drug A is dosed once monthly com-
pared to Drug B, which is weekly.
• Does the route of administration vary be-
tween drugs?
• Determine any potential advantages or dis-
advantages of Drug A versus Drug B.
Step 4: Categorize the Quality of Evidence
Determine the level of evidence for each trial,
including the major and minor limitations
identified.
• Article #1 is a Level I trial with the major
limitation that the two groups are not simi-
lar after randomization, which appears to
have affected the results.
• Article #2 is a Level I trial with minor limi-
tations.
• Article #3 is a Level V trial with major limi-
tations.
136 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Level of Major/Minor
Article Support/Refute Evidence Limitations
1 S I Major-
Randomization
that discredited
the results
2 S I Minor
3 S V Major
Figur
Figuree 9.2: Summary Table of Evidence Evaluated for
Example 9.1 – Individual Patient Decision
Step 5: Develop a Conclusion and Recommen-
dation
Develop a conclusion regarding the overall ef-
ficacy based on the evidence.
• Drug A is equivalent to Drug B in overall
efficacy.
Develop a conclusion regarding the overall
safety based on the evidence.
• Drugs A and B have similar safety profiles.
Review patient’s allergies and potential drug–
drug or drug–disease interactions to limit ad-
verse events.
• No drug–drug interactions were identified
with current therapy.
Develop a conclusion regarding any special
considerations/special populations.
• Drug A is administered significantly less
frequently than Drug B.
Develop a conclusion regarding the overall cost
of medication specific to this individual pa-
tient based on the following:
Chapter 99: Applications 137
•Can the patient afford this medication?
- Insurance coverage
BP’s insurance covers Drug A.
- Patient assistance programs available
• Are there other cost concerns associated
with the treatment?
• Are there long-term cost-saving benefits
of Drug A or Drug B?
Assign the appropriate grade of recommen-
dation based on the quality of evidence avail-
able (Figur
(Figuree 9.3)
9.3).
Supported by ARTICLE
GRADEAA
GRADE
Level I
RECOMMENDATION
RECOMMENDATION
Evidence
Supported by
GRADE AB
GRADE
Level II
RECOMMENDATION
RECOMMENDATION
Evidence
Supported by
GRADEAC
GRADE
Level III, IV, V
RECOMMENDATION
RECOMMENDATION
Evidence
Figur
Figuree 9.3: This Grade of Recommendation Table Is Used to Develop
Each Grade of Recommendation by Tracking Articles and Levels of
Evidence (also located in the “EBM Tool Kit”)
Develop a recommendation based on conclusions
made and applied to this individual patient.
Example:
I strongly recommend using Drug A for the
treatment of osteoporosis in BP for the follow-
ing reasons: (Grade A Recommendation)
• Efficacy – A Level I trial showed that Drugs
A and B were equivalent in efficacy for
treating male osteoporosis. A second Level
138 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
I trial showed that Drug B was superior to
Drug A in the treatment of male osteoporo-
sis; however, this trial had a major limita-
tion that discredited the results. As addi-
tional support, a case series noted an im-
provement in the bone density test after 1
year of receiving Drug A.
• Safety – There appeared to be no differ-
ence in adverse drug reactions between
Drugs A and B. No drug–drug interactions
were noted with BP’s current medication
regimen. BP has no known drug allergies;
therefore, it’s not an issue.
• Special considerations/special populations
– Drug A would allow the patient once
monthly dosing, thus possibly increasing
compliance.
• Cost – Drugs A and B are similar in price.
Drug A will be a less expensive choice for
BP since his prescription insurance cover-
age includes Drug A.
Example 9.2. P opulation-Based
Population-Based
Decision/R ecommendation
Decision/Recommendation
This example illustrates how to compile a class
review or comparative review to assist in the for-
mulary management decision making process.
Step 1: Define the Clinical Question
Determine the actual question being asked so
that an appropriate response is formed. For
this example, the question would be “Is there
a specific drug(s) that is superior to the oth-
ers within the class?”
Chapter 99: Applications 139
Step 2: Retrieve Pertinent Information
Become familiar with class by reading tertiary
textbook resources, compendia class summa-
ries, recent review articles, or class reviews.
Identify medications to be compared and iden-
tify FDA-approved indications.
Complete a literature search for Level I or II
comparative trials by searching various data-
bases. Using the “search and sift” technique,
narrow the search to randomized, controlled
trials with or without power met in human
subjects. In order to ensure a thorough search
is accomplished, be sure to complete a biblio-
graphic search of all relevant articles.
• The search should compare drugs in the
same class, not drugs compared to other
classes or the drug compared to placebo,
if possible.
• If head-to-head comparison trials do not
exist, then consider using placebo-controlled
trials or trials comparing the class to the same
drug that maybe is outside of the class.
Step 3: Evaluate Literatur
Literaturee
Efficacy
• Review each trial and determine if the ar-
ticle supports or refutes the use of one drug
over another.
• Establish if each trial has major or minor
limitations.
Safety
• Assess safety of the drugs within the class
when compared in the literature.
140 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
• Obtain the most recent package inserts to
help determine possible safety concerns;
safety information should always be de-
rived from package inserts rather than in-
dividual trials. Larger patient population
exposure and possibly longer patient ex-
posure are found in package inserts.
Special considerations/special populations
• Establish if one drug is dosed more or less
conveniently than the other(s).
• Does the route of administration vary be-
tween drugs?
• Determine if there are other potential ad-
vantages or disadvantages of the drugs
within the class.
Cost
• Is the cost significantly lower for one drug?
• Are there long-term cost-saving benefits?
• When formulating a recommendation for a
population, fewer specifics are known re-
garding prescription coverage, patient in-
come, and other variables; therefore, cost is
typically one of the final components to con-
sider when forming the recommendation.
• Identify differentiating characteristics for
medications within the class by indication
and use to aid in formulating evidence-
based conclusion.
Step 4: Categorize the Quality of Evidence
Determine the level of evidence for each trial,
including the major or minor limitations iden-
tified.
Chapter 99: Applications 141
Step 5: Develop a Conclusion and Recommen-
dation (see Figur
Figuree 9.2)
Develop a conclusion regarding the overall
efficacy based on the evidence.
Develop a conclusion regarding the overall
safety based on the evidence.
Develop a conclusion regarding any special
considerations/special populations.
Develop a conclusion regarding the overall cost
of medication specific to this population on
the following:
• Cost of Drug X versus Drug Y
• Long-term cost benefits of either drug
Assign the appropriate grade of recommen-
dation (Figure 9.3).
Develop a recommendation statement based
on conclusions and grade of recommendation
to be applied to this population.
Formulate evidence-based recommendation
statement with appropriate justification:
• Efficacy
• Safety
• Special considerations/special populations
• Cost
Example 9.3. P opulation-Based
Population-Based
Decision/R ecommendation
Decision/Recommendation
This example illustrates how to compile a com-
parative review to assist in the formulary manage-
ment decision making process.
142 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
A physician has requested the addition of Drug X
to an institution’s formulary to replace Drug Y for
treatment of chronic obstructive pulmonary dis-
ease (COPD). The pharmacist has been asked to
complete a comparative review of the two agents.
In addition, an evidence-based medicine recom-
mendation needs to be developed that the phar-
macist can present and defend at the next P&T
meeting for the institution.
Step 1: Define the Clinical Question
Determine the actual question being asked so
that an appropriate response is formed. For
this example, the question would be “Should
Drug X replace Drug Y for COPD on the for-
mulary?”
Step 2: Retrieve Pertinent Information
A search of the literature revealed the following
evidence:
Article #1 – A randomized, placebo-controlled
trial was conducted using Drug X in patients
with COPD. Power was not met, thus indicat-
ing that an adequate sample size was not used
to show a difference between study groups, if
a difference really exists. None of the other
ten major considerations were discussed. Pa-
tients receiving Drug X showed greater im-
provement in trough FEV 1 over placebo
(p<0.01). These same patients also showed sig-
nificant improvements in other outcome mea-
sures such as peak expiratory flow rate, de-
creased exacerbations, and health status
scores. The only adverse effect that was noted
Chapter 99: Applications 143
to occur more frequently in the Drug X group
was dry mouth. Patients receiving Drug X who
also had impaired renal function were required
to have dose frequency adjustments.
Article #2 – A randomized, placebo-controlled
trial was conducted using Drug X in patients
with COPD. Power was set but not met, thus
indicating that an adequate sample size was
not used to show a difference between study
groups, if a difference really exists. No other
major limitations were noted. The patients re-
ceiving Drug X showed greater improvement
from baseline than Drug Y in both trough FEV1
(p<0.001) and FVC (p<0.05) and maintained
these effects throughout the study period. Pa-
tients receiving Drug X also experienced sig-
nificantly greater improvement over Drug Y
in peak expiratory flow rate (p<0.01). More
patients in the Drug X group experienced dry
mouth effects than those in the Drug Y group.
Due to the strict inclusion/exclusion criteria,
no special considerations/special populations
were identified. Drug X is more expensive than
Drug Y. Patients had more difficulty using the
drug dispensing device for Drug X versus Drug
Y.
Article #3 – A case report described 12 pa-
tients receiving Drug X who also experienced
dry mouth. These patients were observed by
their family physician who prescribed the drug
to treat their COPD symptoms. No details were
given.
Step 3: Evaluate Literatur
Literaturee
Efficacy
144 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
• Review each trial and determine if the ar-
ticle supports or refutes the use of Drug X
over Drug Y.
- Article #1 – The results of the study
showed that Drug X was superior to
placebo, improving pulmonary func-
tion in COPD patients. Specifically,
patients receiving Drug X showed
greater improvement in trough FEV1
over placebo (p<0.01). These same
patients also showed significant im-
provements in other outcome mea-
sures such as peak expiratory flow rate,
decreased exacerbations, and health
status scores.
- Article #2 – The results of the study
showed that Drug X was superior to
Drug Y, improving pulmonary func-
tion in COPD patients. Specifically,
patients receiving Drug X showed
greater improvement from baseline
than Drug Y in both trough FEV 1
(p<0.001) and FVC (p<0.05) and
maintained these effects throughout
the study period. Patients receiving
Drug X also experienced significantly
greater improvement over Drug Y in
peak expiratory flow rate (p<0.01).
- Article #3 – No details were given re-
garding efficacy in this case report dis-
cussing the safety of Drug X in 12 pa-
tients.
• Establish if each trial has major or minor
limitations.
- Article #1 – Power was set, but not
Chapter 99: Applications 145
met. That is, an adequate sample size
was not used to show a difference be-
tween study groups, if a difference re-
ally exists. However, because there was
a statistically significant difference
noted with the primary endpoint for
Drug X compared to placebo, this is
less of a concern. None of the other
ten major considerations were dis-
cussed.
- Article #2 – Power was set, but not
met. That is, an adequate sample size
was not used to show a difference be-
tween study groups, if a difference re-
ally exists. However, because there was
a statistically significant difference
noted with the primary endpoint for
Drug X compared to placebo, this is
less of a concern. None of the other
ten major limitations were identified.
- Article #3 – This was a case report, and,
therefore did not have a comparative
group. Due to this, there were several
major limitations.
Safety
• Assess safety of Drug X versus Drug Y
when compared in the literature.
- Article #1 – The only adverse effect that
was noted to occur more frequently in
the Drug X group was dry mouth.
There was no difference in the inci-
dence or severity of adverse drug re-
actions between the two drugs.
- Article #2 – Dry mouth was the only
adverse effect noted to occur more fre-
146 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
quently in the Drug X group compared
to Drug Y.
- Article #3 – This was a case report de-
scribing 12 patients receiving Drug X
that also experienced dry mouth.
• Retrieve the most current prescribing in-
formation to determine possible safety
concerns.
- Dry mouth is the most frequent ad-
verse effect associated with Drug X.
Special considerations/special populations
• Establish if Drug X dosed more or less con-
veniently than Drug Y.
- Same dosing frequency
• Does the route of administration vary be-
tween drugs?
- Patients had more difficulty using the
drug dispensing device for Drug X ver-
sus Drug Y.
• Determine if there are other potential ad-
vantages or disadvantages of Drug X ver-
sus Drug Y.
• Are there any other specific situations that
may affect the population positively or
negatively?
- Patients receiving Drug X who also had
impaired renal function were required
to have dose frequency adjustments.
Cost
• Is the cost of Drug X significantly higher
than Drug Y?
- Drug X is more expensive than Drug
Y.
Chapter 99: Applications 147
• Are there long-term cost-saving benefits
of Drug X or Drug Y?
• When formulating a recommendation for
a population, practitioners have fewer
known specifics regarding prescription
coverage, patient income, and other vari-
ables; therefore, cost does not weigh as
heavily and is typically one of the final
components taken into consideration
when forming the recommendation.
Step 4: Categorize the Quality of Evidence
Determine the level of evidence for each trial:
• Article #1 is a Level II trial with major limi-
tation of power.
• Article #2 is a Level II trial with major limi-
tation of power.
• Article #3 is a Level V trial with major limi-
tations.
Level of Major/Minor
Article Support/Refute Evidence Limitations
1 S II Major-Power
2 S II Major-Power
3 Neither support V Major
or refute
Figur
Figuree 9.4: Summary Table of Evidence Evaluated for
Example 9.3 – Population-Based Decision
148 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Step 5: Develop a Conclusion and Recommen-
dation (see Figur
Figuree 9.4)
Develop a conclusion regarding the overall
efficacy based on the evidence.
• Drug X is superior in efficacy to Drug Y.
Develop a conclusion regarding the overall
safety based on the evidence.
• The frequency of dry mouth occurring was
greater with Drug X than Drug Y.
Develop a conclusion regarding any special
considerations/special populations.
• The dispensing device was more difficult
to use for Drug X than Drug Y.
• Patients with impaired renal function re-
quire dose frequency adjustments with
Drug X.
Develop a conclusion regarding the overall cost
of medication specific to this population based
on the following:
• Cost of Drug X versus Drug Y
- Drug X is more expensive.
• Long-term cost benefits of either drug
Assign the appropriate grade of recommen-
dation (Figure 9.3).
Develop a recommendation statement based
on conclusions made and applied to this popu-
lation.
Example:
Recommend replacing Drug Y with Drug X
on the institution’s formulary for treatment of
COPD for the following reasons: (Grade B Rec-
ommendation)
Chapter 99: Applications 149
• Efficacy – A Level II trial shows that Drug
X is superior to placebo for improving
pulmonary function in COPD patients. In
addition, a Level II trial shows Drug X is
superior to Drug Y in improving pulmo-
nary function in COPD patients.
• Safety – With the exception of dry mouth,
there appears to be no difference in ad-
verse drug reactions between Drug X and
Drug Y.
• Special considerations/special populations
– In renal impaired patients, dosing fre-
quency adjustment is necessary with Drug
X in patients with COPD. Patients who
may have difficulty handling the drug dis-
pensing device for Drug X should consider
treatment with Drug Y to ensure treatment
is being administered properly.
• Cost – Drug X is more expensive than Drug
Y; however, there is superior improvement
in pulmonary function with Drug X that
justifies the additional cost. This improve-
ment in pulmonary function may lead to
fewer emergency department visits and
fewer hospitalizations, therefore, lowering
overall cost.
Summary
Phamacy practitioners make both individual patient and popu-
lation-based decisions on a regular basis. It is imperative that
they apply the 5-Step Evidence-Based Medicine Process to en-
sure the highest quality and most reliable evidence is being used
in their clinical decision making. In addition, practitioners need
to consider efficacy, safety, special considerations/special popula-
150 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
tions, and cost when making a recommendation. By taking a less
conservative approach with individual patient decisions and a
more conservative approach with population decisions, the phar-
macy practitioner can be assured that the appropriate interven-
tion or treatment is being recommended.
Chapter 10
Applying EBM
Principles to
Dietary Supplement
Therapeutic
Decisions
Cydney E. McQueen & Celtina K. Reinert
EBM Principles Applied to Dietary Supplements
Differences in Process for Dietary Supplements
Step 2 – Retrieve Pertinent Information
Multiple common names
Plant taxonomy changes
Alternate spellings and misspellings
Step 3 – Evaluate Literature
Power and/or sample size
Biostatistics
Product/dose
Blinding
Outcome measures
Step 5 – Develop a Conclusion and Recommendation
Summary
References
EBM Principles Applied to Dietary Supplements
Due to the increased use of dietary supplements over the past
decade, health care providers are encountering patients who use
151
152 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
supplements more often than they did previously. The pharma-
cist is frequently asked to make recommendations regarding the
therapeutic use of supplements because these products are sold
in many pharmacies. Just as with questions about the use of pre-
scription drugs, the most reliable method of making recommen-
dations is by using evidence-based medicine processes.
The same general principles of obtaining information that
have already been discussed also apply to dietary supplements:
When information is needed, resources are searched in
the same order: tertiary, secondary, and then primary.
Information found is verified in more than one source.
Clinical knowledge and judgment are used along with
the factual evidence, especially in making decisions about
individual patient care.
When a decision must be made based on primary literature,
the same 5-Step Evidence-Based Medicine Process applies to di-
etary supplements.
Figur
iguree 10.1: 5-Step Evidence-Based Medicine Process
Differences in 5-Step Evidence-Based Medicine
Process for Dietary Supplements
If all of these principles and processes are the same, why does
this book have an entire chapter on the topic of using evidence-
based decision making for dietary supplements? Because three of
the EBM process steps may have crucial differences:
Step 2 – Retrieve Pertinent Information: Different search
techniques and sometimes different tertiary and second-
ary resources must be used to ensure that retrieval of pri-
mary literature is thorough.
Chapter 10
10: Applying EBM Principles to Dietary Supplement Therapeutic Decisions 153
Step 3 – Evaluate Literature: Different issues arise in the
analysis of clinical trial quality.
Step 5 – Develop a Conclusion and Recommendation:
Different issues apply in making recommendations.
Step 1 (Define the Clinical Question) and Step 4 (Categorize
the Quality of Evidence) do not differ in any way when deal-
ing with decisions about dietary supplement therapies, so only
Steps 2, 3, and 5 are discussed here.
Step 2 – Retrieve Pertinent Information
In drug information practice, many resources are accepted as
“gold standards.” That term means professionals place a very
high degree of trust in the information within the reference.
For instance, USP DI and AHFS Drug Information are gold
standard drug information resources. Although it is always a
good idea to verify information using another gold standard
reference, that verification is normally done as a simple re-
flection of good clinical practice—pharmacists double-check
everything! Errors caught by this practice are usually on the
level of simple typographical or note transcription errors.
In the area of dietary supplements, no references are con-
sidered “gold standard.” However, some are gaining promi-
nence; see the table in Figur
Figuree 10
10..1 . Every reference pub-
lished to date has flaws or expresses conclusions and recom-
mendations that vary from other recommended resources.
The same studies and case reports are often cited for con-
flicting or even directly opposing recommendations. In other
words, the factual scientific evidence used by the authors to
make a recommendation is generally the same, but the clini-
cal interpretation and judgment allow for different conclu-
sions.
154 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Online R esour
Resour ces
esources
Approx- Type of
Approx- Strengths
Strengths W eaknesses
Weaknesses
imate Information or or
Name Cost Contained Advantages Disadvantages
Natural Medicine $$ General One of the most Discussion given is
Comprehensive comprehensive re- very brief, and all
Database1 sources available. background infor-
www.naturaldata- Direct and to-the- mation necessary
base.com point, each state- to make informed
ment is referenced; decisions may not
hyperlinks to be given.
PubMed citations
and abstracts are
provided. Updated
regularly and
quickly when new
information be-
comes available.
Can also research
interactions, look
up products by
brand names, and
search by medical
condition.
Natural Standard2 $$$$ General Contains detailed Smaller number of
www.natural- information on supplements dis-
standard.com common dietary cussed due to de-
supplements. Ex- tailed process re-
tensive critical lit- quired for mono-
erature review is graph develop-
provided in the ment. Natural
Professional Mono- Standard is cur-
graphs. Direct links rently only avail-
provided to able for purchase
PubMed abstracts. by university and
Bottom-line mono- college level aca-
graphs provide the demic institutions,
same information as medical institu-
the Professional tions such as hos-
Monographs with- pitals, and phar-
out the extensive macies.
literature review.
Monographs orga-
nized by medical
Figur
iguree 10.1: Recommended Dietary Supplement Information Resources
Chapter 10
10: Applying EBM Principles to Dietary Supplement Therapeutic Decisions 155
Appr ox-
Approx- Type of Strengths
Strengths W eaknesses
Weaknesses
imate Information or or
Name Cost Contained Advantages Disadvantages
conditions and
uses. An interac-
tion identifier is
available. Supple-
ments are indexed
by brand name.
Consumer- $ Product quality, Independent labo- Limited scope of
Lab.com3 General ratory that tests product quality.
www.consumer- products for
lab.com amount of product
compared to the
claimed amount,
purity and tablet
dissolution or dis-
integration. The
Natural Products
Encyclopedia, a
general resource,
is also held within
this site.
Print R esour
Resour ces
esources
Natural Medi- $$ General Contains mono- Only updated once
cine Comprehen- graphs in the same yearly, when new
sive Database4, format as online book is published.
published version. Also con- Dated info, time-
yearly tains reference list lag. No interaction
ISBN: and some brand check as database.
0978820517 name products and
content.
Natural Stan- $$$ General Same as the Profes- Not updated as
dard Herb & sional Monographs regularly as the da-
Supplement from the online tabase. Extra tools
Reference: version. provided online are
Evidence-Based not included.
Clinical Reviews5
(2005)
ISBN:
0323029949
156 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Approx- Type of
Approx- Strengths
Strengths W eaknesses
Weaknesses
imate Information or or
Name Cost Contained Advantages Disadvantages
Natural Standard $ General Provides same in- Not updated as
Herb & Supple- formation as bot- regularly as the
ment Handbook: tom-line mono- database. Extra
The Clinical Bot- graphs from the tools provided
tom Line6 (2005) online version. online are not in-
ISBN: cluded.
0323029930
Professional’s $ General Monographs in a Minimal discus-
Handbook of short, quick, easy- sion of studies and
Complementary to-use format. references that
& Alternative monographs are
Medicines, 3rd based on; however,
edition7 (2004) monographs are
ISBN: based on quality
1582552436 information and
studies available.
Herb $ Interaction, Provides known, Only provides in-
Contraindications Contrain- well-documented teraction and con-
& Drug Interac- dications interactions and traindication in-
tions, 3rd edition8 contraindications formation. Lim-
(2001) as well as theoreti- ited to botanical
ISBN: cal ones. Safety- supplements only.
1888483113 oriented resource.
The 5-Minute $$ General Facing pages pro- Stylistically is dif-
Herb & Dietary vide concise infor- ferent from other
Supplement Con- mation on supple- resources, but still
sult9 (2003) ments. user friendly.
ISBN:
0683302736
Evidence-based $ General Contains general Limited number
Herbal Medi- monographs on of monographs,
cine10 (2002) just botanical limited to botani-
ISBN: supplement. Con- cals only.
1560534478 tains some info on
Chinese herbs.
Chapter 10
10: Applying EBM Principles to Dietary Supplement Therapeutic Decisions 157
Approx- Type of
Approx- Strengths
Strengths W eaknesses
Weaknesses
imate Information or or
Name Cost Contained Advantages Disadvantages
Botanical Medi- $$ General, Monographs are Small number of
cine: The Desk Pregnancy focused on the products covered,
Reference for chemistry of the not a user-friendly
Major Herbal supplements. resource in a clini-
Supplements, 2nd Most thorough ref- cal setting due to
edition11 (2002) erence available in focus on chemis-
ISBN: regards to preg- try rather than
0789012669 nancy information. clinically impor-
tant information.
Herbal Medi- $$$ General Partner herbal British publisher,
cines, 3rd edi- book to Dietary language and style
tion12 (2007) Supplements; same differences
ISBN: publisher, basic
0853696233 monograph.
Dietary Supple- $$ General Partner dietary British publisher,
ments, 3rd edi- supplement book language and style
tion13 (2007) to Herbal Medi- differences
ISBN: cines; same pub-
0853696535 lisher, basic mono-
graph.
158 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Example 10.1. Differ ent R
Different esour
Resour ces,
esources,
Differ ent Answers
Different
Three practitioners are asked to answer the ques-
tion, “Can ginger be used in pregnancy and, if so,
at what dose?” If each person was given a differ-
ent resource to use, each one would come up with
a different answer.1,6,7
Natural Medicines Compr ehensive Database:
Comprehensive
“Possibly Safe” and “Possibly Effective;” 250 mg
QID
Pr ofessional’s Handbook of Complementar
Professional’ Complementaryy &
Alter native Medicines: “Use in pregnancy is con-
Alternative
traindicated”
Natural Standar
Standardd Herb & Supplement Hand-
book: The Clinical Bottom Line: Safe and effec-
tive; 1–2 g daily in divided doses for 1–5 days
For these reasons, all information, either factual or interpreted
from the facts, should be verified in a minimum of three resources.
If three references have conflicting information or fail to offer a
clear consensus, more resources should be used (see Example
10.1
10.1). Primary literature may also need to be utilized in addition
to tertiary references.
If three references have conflicting information
Key or fail to offer a clear consensus,
Idea more resources should be used.
Generally, tertiary resources for prescription drug products
include information on interactions, adverse events and
contraindications based on reports from clinical studies, post-
marketing surveillance data, and sometimes published case reports.
Chapter 10
10: Applying EBM Principles to Dietary Supplement Therapeutic Decisions 159
This is true, especially if there are multiple reports of a particular
problem. Most dietary supplement references do not include a cau-
tion against use in a particular disease if that caution is based only
on a theoretical mechanism of action and if no clinical reports sup-
port the concern as significant. Why? Two reasons are cited: 1)
there is far less clinical research on supplements than non-supple-
ments and 2) reporting of problems with supplements is extremely
low. In addition, post-marketing surveillance data are non-exis-
tent. In order to “err on the side of caution” for patient protection,
practitioners need to be aware of theoretical interactions,
contraindications, and potential adverse effects (see Example 10.2
10.2).
Example 10.2. Differ ent Evidence,
Different
Differ ent R
Different ecommendations
Recommendations
A practitioner has a diabetic patient who would
like to try an herbal product for mild osteoarthri-
tis. The two columns represent two different situ-
ations based on the evidence available.
SITUA TION A
SITUATION SITUA TION B
SITUATION
References list a possible References list a possible
side effect of “decrease in side effect of “decrease in
blood sugar.” blood sugar.”
Several clinical trials and Decrease in blood sugar
two published case re- is extrapolated from
ports have documented knowledge of a mecha-
patients experiencing nism of action; no de-
hypoglycemic effects creases have been re-
while using the herb. ported in any clinical tri-
als or case reports.
Recommendation: ad- Recommendation: advise
vise patient to avoid us- patient of possible effect,
ing this herb. titrate up the dose of herb,
and have patient monitor
blood sugar more closely
for 1 or 2 weeks.
160 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
In the United States, most health care practitioners depend
upon Medline to electronically search for clinical trials, case re-
ports, or other articles about many medical topics. For dietary
supplement information, use of Medline alone is not sufficient to
be assured of a thorough search for primary literature. Several
journals that tend to publish more dietary supplement literature
are not indexed in Medline. Many of them are indexed in EMBASE,
another major electronic secondary resource. About one third of
EMBASE-indexed journals are in common with Medline; how-
ever, EMBASE indexes more European and Asian journals. Euro-
pean researchers generally conduct more trials in the area of di-
etary supplements; therefore, they often publish in the European-
based journals. Trials of traditional Oriental herbal medicines or
other complementary and alternative medical therapies such as
qi gong or acupuncture are often found in Asian journals.
EMBASE, in addition to other resources identified in Figure
10.1, can sometimes be found in drug information centers and/
or libraries associated with academic medical centers. Some elec-
tronic resources identified are highly specialized, and locating
facilities for access may be difficult. Unfortunately, many health
care practitioners outside of academic or research settings will
not have access to these resources, so maximizing search results
via other methods becomes even more important.
Bibliographic searching involves scanning the citation list of
trials, review articles, and meta-analyses to find other primary
literature. This is a good method of expanding one’s “reach” for
searching since authors may have access to other indexing sys-
tems and, therefore, have been able to locate additional articles.
Using multiple indexing systems will not ensure a thorough
search if appropriate search terms are not used. Unfortunately,
identifying appropriate search terms is not a simple process. In-
dexing terms for supplements, especially botanical supplements,
differ among indexing systems and change over time.
Common issues with indexing of dietary supplements are
the following:
Multiple common names
Chapter 10
10: Applying EBM Principles to Dietary Supplement Therapeutic Decisions 161
Plant taxonomy changes
Alternative spellings and misspellings
Multiple common names
Many supplements have multiple common names, so a thorough
search should at least include the most frequently used names.
For instance, co-enzyme Q10 is also referred to as ubiquinone. A
recent search of Medline using the medical subject headings
(MeSH) term, ubiquinone, returned 4,592 results, while a key-
word search of “co-enzyme Q10” returned 27 results, 9 of which
were not contained in the MeSH term search.
With botanical supplements, a common problem is that dif-
ferent species may have the same common name. For instance,
two unrelated plants may each be referred to as yellowroot: Hy-
drastis canadensis (usually known as goldenseal) and Coptis
chinensis (usually known as goldthread).
Plant taxonomy changes
As in any scientific field, new knowledge changes things. Plants
are sometimes reclassified and given new scientific names as bota-
nists learn more about them. When this happens, cross-refer-
encing of the old and new names does not always occur, so search-
ing both is necessary. For instance, the current scientific name of
black cohosh is Actaea racemosa, but many resources still refer to
it by the former name, Cimicifuga racemosa. Often newer resources
and electronic databases will include both current and former
names.
Alternate spellings and misspellings
“Ginko” is an older spelling of “ginkgo.” When searched as a
keyword in Medline, “ginko” returns 10 articles that are not lo-
cated using a keyword search of “ginkgo.” A search of “liquorice”
returns 269 articles not found when searching the American spell-
ing of “licorice.” Searching both names locates 351 articles that
are not found when searching the MeSH term for the licorice
family, “Glycyrrhiza.”
At times, searching the same term as both a keyword and a
MeSH term will provide different results. Searching with the sci-
162 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
entific name for feverfew, Tanacetum parthenium, as a keyword
located 28 more articles than searching Tanacetum parthenium as
a MeSH term.
To ensure thoroughness, searches must use common names,
different spellings, scientific names, and alternate scientific names.
The search should be conducted via multiple methods.
In order to ensure a thorough search for primary
Key
literature on dietary supplements, multiple search
Idea methods and alternative terms must be used.
Step 3 – Evaluate Literature
Once the search for primary literature has led to retrieval of clini-
cal trials, the next step is to evaluate those trials. The evaluation
process is identical to the one used to analyze the quality of pre-
scription drug products as outlined in Chapter 5: Evaluate Lit-
erature, but a few different issues arise.
A common complaint about trials of dietary supplements and
other alternative therapies is that they are of poor quality. Al-
though it is not always true, dietary supplement trials are often
small or moderate in size and have significant design limitations.
First, limited funding is available for research in this area. Gov-
ernment agencies such as the National Institute of Health’s Na-
tional Center for Complementary and Alternative Medicine are
providing more funding for large, professionally managed stud-
ies. Many trials are still conducted on a shoestring budget by
practitioners who are not properly trained in how to conduct
research. These disadvantages can sometimes affect the overall
quality of study design. Another problem is that the researchers
may be experts in their field of practice, but lack expertise in use
of supplements or natural therapies. Because there may be mul-
tiple problems with these trials, evaluation of evidence quality
and systematic decision making becomes even more important.
Chapter 10
10: Applying EBM Principles to Dietary Supplement Therapeutic Decisions 163
Several trial limitations common to dietary supplement trials
have been identified:
Power and/or sample size
Biostatistics
Product/dose
Blinding
Outcome measures
Power and/or sample size
The smaller sample size often observed with dietary supplement
trials is primarily an issue of funding. Many supplement manu-
facturers interested in conducting research are simply not large
or profitable enough to afford to enroll a substantial number of
patients to confidently determine differences between groups.
Biostatistics
This limitation could also be related to lack of funding (i.e., not
being able to afford statistician consultation). In addition, research-
ers could lack expertise. Today, it is very easy for the average
person to buy and use statistical programs to analyze data … and
very easy to tell the computer to use the wrong test!
Product/dose
The best dose of a dietary supplement is frequently not known
and doses for trials are based on common usage, which may or
may not be an effective dose (see Example 10.3
10.3).
Example 10.3. Differ ent P
Different Prreparations,
Differ ent Unknowns
Different
There are multiple species of echinacea, three of
which are used medicinally. To date, it is unknown
which of the three species is the most effective,
which plant parts are best used, or what is the best
type of preparation (whole herb, pressed juice, ex-
tract). These “unknowns” may help to explain the
highly variable results seen in efficacy trials.
164 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
A common complaint by medical herbalists or naturopaths
is that researchers often use inappropriate products or prepara-
tions. For instance, a study of glucosamine hydrochloride does
nothing to answer the clinical question about whether glu-
cosamine sulfate relieves symptoms of osteoarthritis (see Example
10.4
10.4).
Example 10.4. Differ
Different ent P
Prroducts,
Differ ent R
Different esults
Results 14,15
Several years ago, while headlines were trumpet-
ing the results of a study saying, “garlic has no
effect on lipid levels,” natural medicine practi-
tioners were replying, “of course it didn’t work!”
Researchers had chosen to use a garlic oil supple-
ment that was heat processed—the high tempera-
tures destroyed all allicin content, the ingredi-
ent believed to be the one responsible for any
positive effects.
Another complaint that has been truer in the past than re-
cently is the use of insufficient doses of comparator drugs. For
instance, many of the early St. John’s wort studies for depression
used only 75 mg of imipramine, a starting dose that would gen-
erally be subtherapeutic for most patients.16 These studies pro-
duced results unfairly biased in favor of the dietary supplement.
Blinding
Blinding difficulties are usually an issue with botanical supple-
ments rather than with the non-botanical supplements such as
melatonin and glucosamine. Many of these plant products have
very distinctive tastes or smells, and it can be impossible to make
identical placebos. For instance, the herb valerian, used for anxi-
ety, sleep, or muscle cramps and spasms, is quite pungent and
identifiable.
Another type of blinding difficulty is when a supplement has
a very distinctive side effect that allows it to be distinguished
Chapter 10
10: Applying EBM Principles to Dietary Supplement Therapeutic Decisions 165
from the placebo. For instance, fenugreek, a supplement tested
for effects on lowering blood sugar, has been reported to cause a
“maple syrup” smell in the urine of many who use it.
Outcome measures
Another limitation often seen in supplement trials is the use of
non-standard outcome measures (i.e., measures not commonly
used and accepted for monitoring symptoms of a disease state).
For instance, if researchers create a questionnaire about symp-
toms of depression rather than using a validated measurement
such as the Hamilton Depression Scale, the results would not be
widely accepted by other medical practitioners.
These areas are not the only ones in which methodological
limitations might be found. They are simply the areas in which it
is most common to find problems when critiquing trials of di-
etary supplement therapies.
Step 5 – Develop a Conclusion and
Recommendation
This step of the process uses the same principles as non-dietary
supplement recommendations described in Chapter 7: Develop
a Conclusion and Recommendation; however, some different fac-
tors need to be considered.
Patient autonomy is a greater issue. Many patients who use
supplements place a higher priority on making decisions about
their health and treatment. Although health care providers have
the duty to provide accurate information and their own recom-
mendations to a patient, the right to self-determination must be
respected.
Health care providers have the duty to provide
Key accurate information and recommendations
Idea to patients; however, patient autonomy
must be respected.
166 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Another concern to keep in mind is product quality. Patients
and practitioners don’t generally have to think about quality with
prescription drugs, but contamination, adulteration, and differ-
ing amounts of claimed active ingredients can be a serious issue
with some supplements. When making recommendations about
using a specific type of supplement, health care providers should
also help guide patients to a high-quality product. Brands rec-
ommended by health care professionals should either be partici-
pating in a quality seal program or be tested by a third-party
laboratory. At the very least, products should be made by a large
reputable company, preferably one that also produces prescrip-
tion or OTC drugs (see the table in Figur
Figuree 10.2
10.2).
Patients and care providers should work together to evaluate
both the known and possible risks and benefits of any particular
supplement when making a therapeutic decision. Some ques-
tions to consider are as follows:
Is this product safe for use?
• Do data show it is generally safe and non-toxic? Is
there information on dose-related toxicities? Consider
if the patient has any specific contraindications to use
of the drug, or if he/she is on any medications that
interact with the supplement.
Is this product effective for use?
• If the product is not effective, how much, if any, harm
will be done by not treating the patient’s condition?
Also, consider the extent of effectiveness. For example,
a patient who needs to lower cholesterol by 20 mg/dL
might be a good candidate for a supplement in addi-
tion to lifestyle changes, while a patient who needs
lowering by 120 mg/dL is not a good candidate.
Is this condition self-treatable and able to be self-moni-
tored?
• Some conditions cannot be considered self-treatable
because of the potential for worsening of the disease.
An example would be depression due to the risk of
suicide.
Chapter 10
10: Applying EBM Principles to Dietary Supplement Therapeutic Decisions 167
Method
of Assurance Definition Examples Comments
Quality Seal If a manufacturer DSVP – Dietary All quality seal programs
Programs agrees to abide by Supplement Verifi- are voluntary and may
standards and cation Program have different standards of
quality control from the United quality.
measures as set by States Pharma-
the offering com- copeia, the stan-
pany or institu- dard-setting orga-
tion, it is allowed nization for
to display a seal United States drug
on supplement la- manufacturers.17
beling as an indi-
cator that the TruLabel – from Mandatory for NPA mem-
product meets the Natural Prod- ber companies that manu-
certain require- ucts Association.18 facture their own labeled
ments. products.
Ratings from A third-party Consumer ConsumerLab also offers a
a Third-Party laboratory pur- Lab.com19 quality seal program.
Laboratory chases supple-
ments “off the Consumer Re- Only reviews products oc-
shelf” and tests ports from Con- casionally, so information
them for quality. sumers Union in magazines may be out of
date. Some information
from the Natural Medicines
Comprehensive Database is
available on the Consumer
Reports web site.
Reputable A large, reputable Glaxo-Smith The rationale behind this
Manufacturer company that also Kline – produces assurance method is that
produces pre- prescription prod- these companies are re-
scription or non- ucts such as quired to follow Good
prescription Amoxil® and Manufacturing Practices
drugs. Zantac®, and also (GMPs) for the drug prod-
the dietary ucts they produce. If they
supplement, already have such strict
Alluna®.20 quality control procedures
in place for some products,
they are more likely to use
quality control measures for
other products.
Figur
iguree 10.2: Product Quality
168 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Is this a high quality product?
• For example, is it in a quality seal program or has it
been evaluated by a third-party program?
Do the benefits outweigh the risks of using the product?
Is there another product (prescription, OTC, or dietary
supplement) that would work better for this patient and
this condition?
Summary
The process of evidence-based decision making for dietary supple-
ments is the same as for prescription drugs, although a few dif-
ferences apply:
Primary literature retrieval search techniques require more
thought and planning, and perhaps more “legwork,” since
more indexing systems must be used.
Literature evaluation generally requires consideration of
different, and sometimes more, limitations than prescrip-
tion drug studies.
Additional considerations need to be addressed when
making recommendations.
References
1. Jellin JM, Gregory PJ, Batz F, et al., eds. Natural Medi-
cines Comprehensive Database [database online]. Stock-
ton, CA: Therapeutic Research Faculty. Available at
www.naturaldatabase.com.
2. Basch E, Ulbricht C, eds. Natural Standard [database
online]. Cambridge, MA; Natural Standard. Available at
www.naturalstandard.com.
3. Cooperman T, Obermeyer W, eds. ConsumerLab.com.
[database online]. White Plains, NY: ConsumerLab.com,
LLC. Available at www.consumer lab.com.
4. Jellin JM, Gregory PJ, Batz F, et al., eds. Pharmacist’s Let-
ter/Prescriber’s Letter Natural Medicines Comprehensive Da-
Chapter 10
10: Applying EBM Principles to Dietary Supplement Therapeutic Decisions 169
tabase. 9th ed. Stockton, CA: Therapeutic Research Fac-
ulty; 2007.
5. Ulbricht CE, Basch EM, eds. Natural Standard Herb &
Supplement Reference: Evidence-Based Clinical Reviews. St.
Louis, MO: Mosby, Inc.; 2005.
6. Basch EM, Ulbricht CE, eds. Natural Standard Herb &
Supplement Handbook: The Clinical Bottom Line. St. Louis,
MO: Mosby, Inc.; 2004.
7. Fetrow CW, Avila JR, eds. Professional’s Handbook of Comple-
mentary & Alternative Medicines. 3rd ed. Philadelphia, PA:
Lippincott; 2004.
8. Brinker F. Herb Contraindications & Drug Interactions. 3rd
ed. Sandy, OR: Eclectic Medical Publications; 2001.
9. Fugh-Berman A. The 5-Minute Herb & Dietary Supplement Con-
sult. Philadelphia, PA: Lippincott, Williams & Wilkins; 2003.
10. Rotblatt M, Ziment I, eds. Evidence-Based Herbal Medi-
cine. Philadelphia, PA: Hanley & Belfus; 2002.
11. McKenna DJ, Jones K, Hughes K. Botanical Medicine: The
Desk References for Major Herbal Supplements. 2nd ed. New
York, NY: The Haworth Herbal Press, Inc; 2002.
12. Barnes J, Anderson LA, Phillipson JD. Herbal Medicines.
3rd ed. London, UK: Pharmaceutical Press; 2007.
13. Mason P. Dietary Supplements. 3rd ed. London, UK: Phar-
maceutical Press; 2007.
14. Berthold HK, Sudhop T, Bergmann K. Effect of a garlic oil
preparation on serum lipoproteins and cholesterol me-
tabolism: a randomized controlled trial. JAMA. 1998;
279(23):1900–1902.
15. Lawson L. Effect of garlic on serum lipids. JAMA. 1998;
280(18):1568.
16. Vorbach EU, Hubner WD, Arnoldt KH. Effectiveness and
tolerance of the hypericum extract LI 160 in comparison
with imipramine: randomized double-blind study with
135 outpatients. J Geriatric Psychiat Neurol. 1994; 7(Suppl
1):S19–23.
170 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
17. United States Pharmacopeia. USP Verified Dietary Supple-
ments [database online]. Available at: http://www.usp.org/
USPVerified/dietarySupplements. Accessed August 2007.
18. Natural Products Association (formerly National Nutri-
tional Foods Association) TruLabel Program [online da-
tabase]. Available at: http://www.naturalproductsassoc.org/
site/PageServer?pagename=ic_bg_trulabel. Accessed Au-
gust 2007.
19. ConsumerLab. The CL Seal. Available at: http://
www.consumerlab.com/seal.asp. Accessed August 2007.
20. GlaxoSmithKline. Product list. Available at: http://
www.gsk.com. Accessed May 2008.
Glossary of
Evidence-Based
Medicine Terms
Absolute Risk Reduction (ARR) – The absolute difference be-
tween the rates of outcomes between two groups. For ex-
ample, let us assume that a study evaluating the efficacy of
two different medications in preventing secondary myocar-
dial infarctions (MI) in patients with known risks was per-
formed using two active treatments. For patients receiving
Treatment A, 15% experienced a secondary MI compared to
only 5% of patients receiving Treatment B who experienced a
secondary MI. The ARR for Treatment B is (A – B) or 0.15 –
0.05, which equals 0.1 or 10%. This value may be more use-
ful in evaluating the true impact of a therapy versus relying
on the relative decrease in risk (see Relative Risk Reduction
and Number Needed to Treat).
Alpha Level ((α α) – The probability of making a Type I error (see
Type I Error). In hypothesis testing, this value is also the p-
value threshold for a result to be statistically significant. Sta-
tistical significance is noted to occur when the p-value is less
than the α-level.
Alter native Hypothesis – The study hypothesis that is usually the
Alternative
exact opposite of the null hypothesis (see Null Hypothesis). In
medical research, the alternative hypothesis usually states that
there is a difference between study groups. For example, if the
null hypothesis is that A = B, then the alternative hypothesis
could be A ≠ B. There can also be multiple alternative hypoth-
eses. Using the previous example, the multiple alternative hy-
potheses could be set up as 1) A > B and 2) B > A.
Applicability – How well the results of a clinical study will fit
into "real world" practice settings. Also referred to as relevance.
171
172 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Bayesian Analysis – An analysis beginning with a specific prob-
ability that then incorporates new data or information result-
ing in a revised probability (prior probability and posterior
probability, respectively).
Beta Level ((ββ) – The probability of making a Type II error (see
Type II Error). This value is related to the “power” of a study,
which equals 1 – β.
Bias – An error credited to the investigative team or subject/patient
that provides results different from the true results and leads to
a potentially wrong interpretation of the overall study results.
Binomial – Data that has only two outcomes.
Blinding – A study design method utilized to neutralize the po-
tential for biased study results based on inaccurate reports
from patients, health care providers, and study investigators.
Classifications of blinding include:
Open Label – No blinding exists. Patients, health care provid-
ers, and study investigators all know what treatment is
being given at all times.
Single Blinding – The patient is unaware of what treatment
arm he/she is assigned. This reduces the potential that a
patient will report a positive or negative outcome based
upon a predisposed “feeling” of how well a treatment
works or does not work (i.e., placebo effect).
Double Blinding – The patient, health care provider, and study
investigators are unaware of what treatment arm the pa-
tient has been assigned. This blinding method reduces
the bias of the patient, health care provider, and study
investigator using predisposed “feelings” when assessing
how a study treatment is working during a clinical trial.
This method is particularly useful because the health care
provider must remain objective in the management of the
patient’s disease state through a clinical trial.
Triple Blinding – The patient, health care provider, study in-
vestigator, and an outcomes assessor are all blinded. In
this scenario, the health care provider is utilized to pro-
vide only data, and a distinctly different individual evalu-
Glossar y 173
ates the data to provide the outcome conclusion. This
individual remains blinded to the assigned treatment arm
in order to maintain objectivity when completing out-
comes assessments. A good example of this situation is
when a patient is being treated for a fungal infection in
the lungs. The health care provider and/or study investi-
gator monitors the patient through the clinical trial. How-
ever, the individual who provides the final determination
of clinical success or failure would be a radiologist exam-
ining the patient’s chest x-rays. Using a very specific set
of criteria, this unbiased, blinded third party makes an
objective determination of clinical success or failure.
Triple blinding is a much less common approach to clini-
cal trial design than double blinding. However, in clinical
trials that rely on more “qualitative” results (e.g., x-rays)
rather than numerical laboratory tests, the use of an in-
dependent third party to determine clinical success or
failure can lead to more scientifically sound results by
eliminating the inter-rater variability present when using
multiple health care providers who might have multiple
opinions of clinical success or failure.
Quadruple Blinding – Very rarely reported, the patient, health
care provider, study investigator, outcomes assessor, and
data analyst are all blinded to the patient’s assigned treat-
ment group.
Case-Contr
Case-Control ol Study – An observational study design method
commonly used when studying rare disease states or out-
comes, such as a rare side effect. A case is identified with the
outcome that the investigator is studying. Control patients
(those lacking the outcome being studied) are then matched
to the case as closely as possible, usually using age, gender,
race, etc. Then, the investigator retrospectively evaluates the
case and control histories to identify any characteristics asso-
ciated with the case group and not associated with the con-
trol group. This method is prone to multiple biases; caution
should be used when making any conclusions based on re-
sults of this trial design.
174 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Case Report – A description of an individual patient with a dis-
ease or outcome that is of interest. There is no control group.
Case Series – A consecutive collection of patients (case reports)
with the same disease that is treated in the same manner.
There is no control group.
Clinical Guidelines – See Practice Guideline.
Clinical Significance – Results from a clinical study that show a
difference large enough to justify changing the way a practi-
tioner would usually treat patients. The results reveal practi-
cal as well as statistical importance.
Cohor
Cohortt – A group of study subjects who share a common expo-
sure or characteristic. A cohort study commonly compares
two large groups of individuals—those who have received
(but not assigned) a specific intervention and those who have
not. The two groups are usually observed prospectively over
time to evaluate the long-term outcomes.
Cointer vention – A treatment or treatments that are not the pri-
Cointervention
mary study intervention (concomitant treatments) that may
actually have an effect that will bias the study results.
Confidence Inter val (CI) – The range or interval of data in which
Interval
the “true” result is thought to exist with a given percentage of
“confidence.” The CI is related to the alpha level set for statis-
tical significance. For example, if the α-level is set at 0.05,
then the CI is 95% (calculated as 1 – 0.05). A 95% CI simply
means that, with 95% confidence, the study results indicate
the true value measured lies within the reported range. How-
ever, there is a residual 5% chance that the true value lies
outside of the calculated range, which prevents a CI from
being a conclusive estimate.
Confounding V ariables – An unknown factor (e.g., patient char-
Variables
acteristic, environmental cause, changes in medical practices
over time) that could have contributed to the effect seen in a
clinical trial, unrelated to the intervention. Researchers aim
to eliminate confounding variables so that the results of a
clinical trial can provide as much conclusive evidence of a
measured benefit as possible. Proper study design is the best
Glossar y 175
way to minimize confounding variables. Randomization of
study subjects to the treatment groups is a common method
of minimizing confounding variables.
Consecutive Sample – A sample of patients, all of whom were
potentially eligible, enrolled, and seen over a period of time.
Contr ol Gr
Control oup – A group of patients that do not receive the
Group
study intervention. They may receive a placebo or other ac-
tive treatment that is often times the "gold standard."
Convenience Sample – A patient or group of patients that are
selected at the convenience of the investigator or were avail-
able at a convenient place or time.
Cor
Corrrelation – The degree of relationship noted between two or
more different variables or observations.
Cor
Corrrelation Coef ficient – A numerical expression (between -1.0
Coefficient
to 1.0) of the degree of relationship noted between two or
more different variables
Cost Analysis – When two or more strategies are analyzed but
only the costs are compared, not costs and expected outcomes.
Cost Benefit Analysis – When two or more strategies undergo
an economic analysis that includes both costs and conse-
quences articulated in monetary terms.
Cost-Ef fectiveness Analysis – When two or more strategies un-
Cost-Effectiveness
dergo an economic analysis that includes both costs and con-
sequences articulated in natural units such as cost per life
saved.
Cost Minimization Analysis – When two or more strategies
undergo an economic analysis where the consequences for
each are the same, thus the only issue is their relative costs.
Crossover T
Crossover rial – A type of study design in which study partici-
Trial
pants receive both treatments. By “crossing over” to the com-
parison treatment arm, each participant serves as its own
control group. When evaluating the outcomes of these stud-
ies, it is important to ensure that there was an appropriate
length of “washout” before the participants crossed over into
the second treatment group. Too short of a washout period
176 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
can allow residual pharmacodynamic effects of the first treat-
ment to influence the results of the second treatment, which
could give misleading results.
Cr oss-Sectional T
Cross-Sectional rial – A type of study design that evaluates a
Trial
defined population at a single point in time. The national
census survey is a good example of a cross-sectional trial.
Decision Analysis – A systematic process using a mathematical
model that includes various identified components to make
the best possible clinical decision.
Decision T Trree – An analytical tool used to express the various
components that go into a decision analysis.
Dependent V ariable – The outcome variable in a study. This
Variable
variable is affected by changes in the independent variable.
Descriptive Statistics – Statistics that summarize, tabulate, and
organize data to describe study observations or measurements.
Effect Size – The difference in outcomes between the study group
Effect
and the control group divided by a measurement of variance
such as the standard deviation.
Effectiveness – The measurement of a treatment’s “true” impact
Effectiveness
on a particular disease state in a “realistic” setting. The effec-
tiveness of a treatment can differ greatly from its own estima-
tion of efficacy (see Efficacy). The effectiveness of a treatment
is influenced by a number of factors, including the frequency
of administration required to achieve a therapeutic effect, the
administration route, the treatment’s cost to the patient, the
treatment’s side effect profile, the patient’s perceived benefit
from the treatment, and the treatment’s social acceptance.
Many of these factors result in patient non-compliance to
therapy. Sometimes, a less efficacious treatment can be more
effective at treating or maintaining a disease state when stud-
ied in the “real world” setting.
Efficacy – The measurement of a treatment’s “potential” impact on
Efficacy
a particular disease state in a “perfect” setting. Efficacy is usu-
ally determined in very controlled situations that do not al-
ways have the same extraneous influences that exist when study-
ing a treatment’s “true” effectiveness (see Effectiveness). Large
Glossar y 177
phase III clinical trials are commonly used to determine a
treatment’s efficacy. The FDA generally approves medications
based on efficacy, not effectiveness, data in addition to other
considerations such as safety. However, once approved and
released to the general population, a medication’s potential effi-
cacy can be very different from its resultant true effectiveness.
Endpoint – Health event(s) or outcome(s) that signify comple-
tion or termination from a trial such as death or loss to fol-
low-up.
Equivalence Study – A study designed to show equivalence be-
tween treatments in safety, efficacy, or other parameters. Also
called non-superiority studies.
Event Rate – Proportion of patients in the experimental group
(experimental event rate) and/or control group (control event
rate) that a predefined event is observed.
Follow-up – A process to determine the outcome in every pa-
tient who participated in a clinical trial.
Frequential Statistics – The statistical method of using frequen-
Frequential
cies of an outcome to determine probabilities or odds of an
outcome occurring. This method does not take into account
past experience with the outcome’s probability, such as a Baye-
sian Analysis method. Frequential statistics are currently the
most common method used to evaluate the results of con-
trolled clinical trials.
Generalizability – The capability to take the results generated
by a study and generalize to a larger and similar population
of patients.
Gold Standar
Standardd – A well accepted therapeutic approach consid-
ered the standard of practice for a particular disease state.
Hawthor
Hawthorne ne Ef fect – The phenomenon where a patient's perfor-
Effect
mance is improved because he/she is aware that his/her be-
havior is being observed.
Hazar
Hazard d Ratio – The weighted relative risk over the time of the
entire study.
178 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Incidence – Number of new cases of a specific disease over a set
time.
Independent V ariable – The treatment variable that is assumed
Variable
to have some effect on the outcome or dependent variable.
Infer ential Statistics – The process of making conclusions based
Inferential
on the results from a sample of the population of interest.
Intention-to-Trreat (ITT) Analysis – A method of statistical analy-
Intention-to-T
sis that maintains randomization by incorporating all patients
in their originally assigned groups during data analysis, re-
gardless of whether the patient finished the study. This also
includes patients lost to followup. This type of analysis can
more accurately depict the clinical significance of a studied
treatment. For example, if the researchers only analyzed the
outcomes of the patients who finished the trial, then the safety
and benefits of a treatment could be over- or underestimated.
ITT Analysis can be very complex in its methods, but a study
that utilizes this analysis generally is considered to have more
validity in its results.
Inter obser
Interobser ver V
observer ariability – Variation in outcome results by dif-
Variability
ferent observers.
Inter val Data – Continuous data scales with known, equal dis-
Interval
tance between each interval (e.g., blood pressure, blood glu-
cose).
Inter ventional Study Design – A study design in which sub-
Interventional
jects receive a treatment (or intervention) they would not oth-
erwise receive and the effect on a particular outcome is ob-
served.
Intraobser ver V
Intraobserver ariability – Variation in outcome results by the
Variability
same observer during repeat testing.
Likert-type Scales – An instrument used by study investigators
to capture a patient's rating of his/her response such as pain;
a scale that measures extremes of attitudes or feelings, gener-
ally using 3 to 9 possible values such as no pain, mild pain,
moderate pain, and severe pain.
Lost to Follow-up – The inability to determine the outcome of a
patient who participated in a clinical trial.
Glossar y 179
Matching – An intentional process to create similar study and
comparative groups based on patient characteristics. This is
seen primarily in observational case-control studies to help
substantiate that differences in results between the groups
are due to the study intervention of interest.
Mean – A measurement of central tendency or, in other words,
the arithmetic average.
Meta-Analysis – A statistical method of compiling results from
various studies to obtain a pooled result representing “new
data.” The goal of a meta-analysis is to increase statistical power
through an increase in the pooled sample size. Meta-analyses
are commonly used to clarify the roles of treatment in spe-
cific clinical situations that have conflicting results. A meta-
analysis can also answer new questions not previously re-
ported in original manuscripts, but this generally requires
access to the original study data that is not often readily avail-
able. It is important to note that a meta-analysis cannot con-
vert a poorly designed study into a well-designed study; the
quality of the results in a meta-analysis is entirely dependent
upon the quality of data going into the analysis.
N of 1 trial – Type of crossover trial in which a single patient or
small numbers of patients serve as their own control and re-
ceive alternating courses of treatment over a specific period
of time.
Nominal Data – Categorical data that cannot be ranked (e.g.,
yes/no, male/female, hair color, eye color).
Non-Dinomial – Data that is not limited to two outcomes (also
non-binomial).
Non-Parametric Distribution – Data that, when graphed, fall
into a non-normal distribution and do not form a bell-shaped
curve.
Non-Parametric T ests – Statistical tests that do not assume a
Tests
normal distribution of data. These statistical tests are used
when the data is skewed, but can also be used for non-skewed
data.
Null Hypothesis – The “baseline” hypothesis that is generally
180 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
thought to be true but has not been proven. In medical re-
search, the null hypothesis usually states that there is no dif-
ference between study groups. There can only be one null
hypothesis in a given study. The directive of the study is to
either reject the null hypothesis OR refuse to reject the null
hypothesis. Note that refusing to reject the null hypothesis is
not the same as accepting the null hypothesis to be true.
Number Needed to Harm (NNH) – The number of patients
who need to receive a treatment before 1 patient is “harmed”
that would not have occurred if all of those patients received
the comparison treatment. NNH is calculated from the recip-
rocal of the absolute percent difference in incident rates of a
specific adverse event between two study groups. For example,
if Treatment A is associated with a 5% incidence of rash, and
Treatment B is associated with a 2.5% incidence of rash, then
the NNH for Treatment A would equal 1 / (0.05–0.025). This
equals 40 patients who need to be treated with Treatment A
before 1 patient gets a rash that would NOT have otherwise
occurred had all 40 patients received Treatment B. Ideally,
the higher the NNH is for a treatment, the better.
Number Needed to T Trreat (NNT) – The number of patients who
need to receive a treatment before 1 patient reports a positive
outcome that would not have occurred if all of those patients
received the comparison treatment. NNT is also equivalent
to the reciprocal of the absolute risk reduction (1 / ARR) (see
Absolute Risk Reduction). For example, if Treatment A is asso-
ciated with a 30-day mortality rate of 20%, and Treatment B
is associated with a 30-day mortality rate of 10%, then the
NNT for Treatment A would equal 1 / (0.2 – 0.1). This equals
10 patients who need to be treated with Treatment A to pre-
vent 1 death at 30 days, which would NOT have otherwise
occurred had all 10 patients received Treatment B. Ideally,
the lower the NNT is for a treatment, the better. Generally,
medications that are used to actively treat a disease state (e.g.,
antibiotics in treating pneumonia) have lower NNTs than
medications that are used to prevent the development of a
Glossar y 181
particular disease state (e.g., aspirin in preventing stroke).
Obser vational Study Design – A study in which no interven-
Observational
tion is made on the subjects; observations are made on natu-
rally occurring events.
Odds – A ratio of probability comparing occurrence to
nonoccurrence of a particular event.
Odds Ratio – The ratio of the odds of exposure in case patients
divided by the odds of exposure in control patients. This is
commonly reported in case-control studies and prospective
cohort studies.
Operator -Dependent – A phenomenon where the results of a
Operator-Dependent
test are dependent upon the specific skills of the person con-
ducting the test.
Ordinal Data – Data that can be ranked in specific order. The
Ordinal
intervals between data points are not equal distance (e.g.,
Likert pain scale).
Outcome – The result(s) associated with a medical intervention
on a subject or patient. Measurements for the outcome are
generally established prior to conducting the trial. Investiga-
tors often establish primary outcome(s) and secondary
outcome(s). The primary outcome(s) is generally used to cal-
culate the required sample size to meet a set power, thus as-
suring that a difference between study groups is identified, if
one truly exists.
P-Value – The probability that random chance is the source of
P-Value
the observed difference between two test variables and is not
related to the treatment being studied. In other words, the
smaller the p-value, the more likely the observed difference
is due to something other than random chance. If the p-value
is less than the pre-established α-level, then the p-value is
considered “statistically significant” (see Alpha Level and Type
I Error).
Parallel Study Design – Study design in which two or more
groups receive treatment simultaneously and are compared.
182 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Parametric Distribution – Data that, when graphed, fall into a
normal distribution of a bell-shaped curve.
Parametric T ests – Statistical tests that assume the data is nor-
Tests
mally distributed (i.e., bell curve).
Per Pr otocol Analysis – A method of statistical analysis where
Protocol
the investigators analyze the outcomes of only those patients
who completed the trial. Patients lost to followup are not
included. This type of analysis can result in overestimating
or underestimating the safety and benefit of a treatment.
PICO – A strategy used to define the clinical question that takes
into consideration the patient(s), intervention(s),
comparison(s), and outcome(s).
Point Estimate – The estimated value from the study sample of
participants. In a representative sample, the point estimate is
considered the best estimate of a population.
Power – The ability of a study to detect a statistically significant
difference between two groups when a difference truly exists.
The lower the power a study has, the more likely it is to find
no statistically significant differences when, in reality, a dif-
ference truly does exist—which is considered a “false nega-
tive” (see Type II Error). Power is directly affected by sample
size, variance, and the magnitude of difference between means.
The determination of a study’s true power can be very com-
plex, but in its most simplistic form power can be calculated
as 1 – β (see Beta Level).
Practice Guideline – A systematically developed series of state-
ments providing guidance and direction for a practitioner
regarding the treatment of a patient's specific disease. These
guidelines are developed by either consensus of experts, evi-
dence-based medicine principles, or a combination of both.
Prevalence – The frequency of a disease determined by the num-
Prevalence
ber of patients in a population who have a specific disease at
a given time.
Prospective Study Design – A study design in which observa-
Prospective
tions are made as the data is collected and groups are fol-
lowed forward in time.
Glossar y 183
Qualitative Resear
Researchch – Research offering insights into behav-
ioral, social, and emotional events associated with health care.
Subjective in nature.
Quantitative Resear ch – Research involving the testing of spe-
Research
cific hypotheses focusing on variables that will provide num-
bers and measurements suitable for statistical analysis. Ob-
jective in nature.
Randomized Contr olled Study – A study design where subjects
Controlled
or patients are randomly assigned to an intervention or a con-
trol group. The randomization is performed to assure that
each subject or patient has an equal opportunity of ending
up in either group(s). When randomization is successful, each
study group is similar in characteristics and demographics.
Ratio Data – Continuous data scales with known, equal distance
between each interval; they contain a non-arbitrary, absolute
zero (i.e., no negative numbers).
Relative Risk – The ratio of the risk of an outcome in a treatment
group divided by the risk of the outcome in the control group.
In a controlled, clinical trial the risk of an outcome is the
same as the probability or incidence. The relative risk of an
outcome is also our “natural” interpretation of risk ratios.
This is commonly the number we consider when describing
a treatment to be “_______ times as likely” to have a particular
outcome.
Relative Risk Reduction (RRR) – The relative difference between
the rates of outcomes between two groups. For example, let
us assume that a study evaluating the efficacy of two different
treatments in preventing secondary myocardial infarctions
(MI) in patients with known risks was performed using two
active treatments. For patients receiving Treatment A, 15%
experienced a secondary MI compared to only 5% of patients
receiving Treatment B who experienced a secondary MI. The
RRR for Treatment B is calculated as (A – B) / A or (0.15 –
0.05) / 0.15, which equals 0.667 or 66.7%. The RRR of a
treatment generally inflates the true benefit of a treatment,
which is why RRR is the most commonly reported number
184 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
in the news media. Absolute risk reduction and number
needed to treat may be better measures to use when compar-
ing two treatment options (see Absolute Risk Reduction and
Number Needed to Treat).
Reliability – The ability of a test or process to provide the same
results when repeated with similar conditions.
Retr ospective Study Design – A study design in which observa-
Retrospective
tions are made looking back in time at data that already exists.
Sample – A part of the population that is selected to participate
in the study.
Sensitivity – A test's ability to identify a true-positive rate such
as the presence of a specific disease.
Sensitivity Analysis – An analytical procedure used to verify what
effects would occur to the results from changing input vari-
ables.
Specificity – A test's ability to identify a true-negative rate such
as the non-presence of a specific disease.
Standar
Standard d Deviation – A measurement of the spread from the
mean. A large variance reflects a large spread from the mean
or, in other words, a large standard deviation.
Statistical Infer ence – See Inferential Statistics.
Inference
Statistical Significance – A measure of confidence concerning
an observed difference between study groups and whether
that difference is due to the study treatment or just chance
alone.
Stratified Randomization – A method to ensure balance between
different study groups regarding important factors that may
have an effect on the outcome.
Sur
Surrrogate Endpoint – A measurable (and usually quantifiable)
test result that makes inferences towards an actual clinical
outcome. Surrogate endpoints are commonly used in clinical
trials to measure the efficacy of a treatment using a smaller
sample size in a shorter amount of time. Also, surrogate end-
points are commonly used in treatments that are in the early
stages of development. As experience is gained with a new
Glossar y 185
treatment, the use of surrogate endpoints is replaced with
actual clinical outcomes data. A surrogate endpoint also gives
patients and health care professionals a biomarker useful for
monitoring the ongoing efficacy of a therapy, such as with
antihypertensive agents and the blood pressure measurement.
Surrogate endpoints must have a strong correlation with a
clinical outcome in order to be useful in making inferences
towards the actual patient outcome. For example, measuring
blood pressure is a good surrogate marker that has substan-
tial clinical data correlating it to the risk of cardiovascular
mortality.
Systematic Review – A formal evaluation involving a pertinent
literature search and critical literature appraisal to answer a
defined clinical question. No "new data" is created.
Treatment Ef fect – A measurement used to express the results of
Effect
comparative clinical studies. These measurements include
Absolute Relative Risk, Relative Risk Reduction, Odds Ratio,
Number Needed to Treat, and Effect Size.
Type I ErErrror – Falsely determining that a statistically significant
difference between two study variables exists when, in real-
ity, no difference truly exists (i.e., false positive). The prob-
ability of a Type I error occurring is related to the alpha level
set by investigators (see Alpha Level).
Type II ErErrror – Falsely determining that no statistically signifi-
cant difference between two study variables exists when, in
reality, a difference truly does exist (i.e., false negative). The
probability of a Type II error occurring is related to the beta
level set by investigators (see Beta Level).
Validity – The measure of confidence one has that the results are
true, believable, and free from bias. Validity can also repre-
sent the level of confidence that a measurement accurately
represents what it is supposed to measure.
Variable – Anything that can take on different values such as an
outcome measurement, characteristic of a group, risk factor, etc.
Variance – A measurement of the spread of values around the
mean. Also see Standard Deviation.
186 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Washout Period – The period of time required for the pharma-
cological effect of a treatment to end once the treatment has
been discontinued. In crossover clinical trials, an adequate
washout period should be maintained between treatment
phases. Failure to do this can result in the pharmacological
effect being carried over to the alternate treatment phase, thus
confounding the results.
Evidence-Based
Medicine Tool Kit
5-Step Evidence-Based Medicine Process
Diagram
Define the Clinical Question
PICO
Initial Patients Defined
Question
Clinical
(no detail Interventions Question
state)
Comparison
- Context Outcomes
- Scope
187
188 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Ten Major Considerations
Ar ticle T
Article itle:_________________________________________
Title:_________________________________________
Level: (circle one)
(circle I II III IV V
Limitations: (circle one)
(circle Major Minor
MAJOR CONSIDERATIONS with Justifications for Each
CONSIDERATIONS S L
Power set/met? yes/no
Dosage/treatment regimen appropriate? yes/no
Length of study appropriate to show effect? yes/no
Inclusion criteria adequate? yes/no
Exclusion criteria adequate? yes/no
Blinding present? yes/no
Randomization resulted in similar groups? yes/no
Biostatistical tests appropriate for type of
yes/no
data analyzed?
Measurement(s) standard/validated/accepted
practice? yes/no
Author's conclusions are supported by the
yes/no
results?
Evidence-Based Medicine Tool Kit 189
Levels of Evidence to Categorize
Quality of Evidence
Control Group
Level I Randomized
Power Met Control Group Interventional
Level II Randomized
confidence
Power Not Met Control Group
Prospective
Level III Randomly Assigned
Observational
Level IV Control Group
Retrospective
Randomly Assigned
Level V Interventional/Observational
No Control Group
Non-Randomized
Summary Table of Evidence Evaluated
Clinical Question -
Level of Major
Article Outcome
Evidence Limitations
190 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Grading Tool to Determine Strength of
Recommendation
GRADE
GRADEAA
Level I TrialsI
LEVEL
GRADE
GRADEBA
Level II Trials
LEVEL I
GRADE
GRADECA
Levels III, IV,
LEVEL I
& V Trials
Recommendation Format
THE RECOMMENDATION STATEMENT
JUSTIFICATION IN A BULLET FORMAT
• Efficacy
• Safety
• Other special considerations/populations
• Cost
Index
A
Absolute risk reduction, 16, 17
Active control trials, 18
Alpha value, 19, 20, 35
Analysis of variance (ANOVA), 34, 35
Appropriate test, data analyzed, 31–32
ATP III (Adult Treatment Panel III), 124
Authority, 2
B
Beta level, 39
Bibliographic searching, 54, 160
Binomial data, 27
Biostatistic concepts, 7–8
appropriate test for data analyzed, 27–
35
dietary supplement studies, 163
power, 21–26
statistical significance, 19–21
Blinding, 18, 74–75, 164–65
Botanical Medicine, 157
C
Case control study, 15
Categorical data, 30, 34–35
191
192 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
Categorize quality of evidence, Consensus guidelines, 124
6, 87–88 Consumer Lab, 155, 167
individual example, 135– Consumers Union, 167
36
Continuous data, 27, 31, 32,
key study attributes, 98– 33, 35
101
Control group, 93
population-based example,
Control trials, 17–18
140, 147
Cook, Deborah, 6, 89
study design and levels of
evidence, 88–90 Cost, 115, 116, 129, 130
study design questions, 91– Crossover interventional trial,
98 12–13
Cautious recommendations, 4
Chi-square test, 33, 34 D
Class review, 138–41
Data, types of, 27–29
Clinical judgment, 25, 108
Decision-making
Clinical Pharmacology, 52, 68,
time-sensitive, 4
69
types of, 108–9, 111
Clinical practice applications,
127–50 Deductive reasoning, 2–3
Clinical question Define clinical question, 5, 41
defining, 41 effects on other evidence-
based medicine activities,
framing with PICO, 42–45
45–48
well-defined, 45
individual decision, 132
Cohort studies, 14
population-based decisions,
Comparative review, 138–41, 138, 142
141–49
problems to avoid, 49
Compendia, 52, 68
tips for, 48–49
Conclusion
using PICO, 42–45
from summary, 105–7
Develop conclusion/recommen-
recommendation from, 107– dation, 6, 103–4
17
conclusion from summary,
Confidence in evidence, 112 105–7
Confidence levels, 104 individual decision, 136–
Conflicting information, 158 38
Index 193
organizing information, population-based decisions,
104–5 139–40, 143–47
population-based decision, Ten Major Considerations,
141, 148–49 62–82
recommendation from Evaluation measurements, 80–
conclusion, 107–17 81
Dietary supplements Evidence
developing conclusion and different recommendations,
recommendation, 153, 165– 159
68 levels of. See Levels of
differences in process for, evidence
152–53 Evidence-based guidelines, 124
EBM principles, 151–52 Evidence-Based Herbal Medicine,
evaluating literature, 153, 156
162–65 Evidence-based medicine
information resources for, defined, 1
154–57
5-step, 5–7, 8. See also
retrieving information, 152, individual steps
153–62
processes, 4, five-step, 5–7,
Dietary Supplements, 157 8
Distribution, data, 29–30 tool kit, 96, 104, 110, 187–
Dosage, appropriate, 67–69 89
Exclusion criteria, 71–74
E not adequate, 73–74
EBM Tool Kit, 96, 104, 110,
187–89 F
Efficacy, 114, 116, 128, 130 Facts and Comparisons, 52
EMBASE, 52, 160 Firm decision, 4
Evaluate literature, 6 Five-Minute Herb & Dietary
dietary supplements, 162– Supplement Consult, 156
65 Formulary management clinical
individual decision, 133– decision, 109
35 Framingham Heart Study, 14
multiple item checklists, 61–
62
194 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
G Interventional randomized
controlled trial, 97–98
Gold standards, 153 Interventional study, 91, 92, 93
Google, 57 look forward, backward,
Grade of Recommendation, 112 94–95
Grade of Recommendation Iowa Drug Information Ser-
Tool, 110 vices, 52
Guyatt, Gordon, 6, 89
J–K
H JNC7 (Seventh Report of Joint
Harvard Nurse’s Health Study National Committee), 124
II, 94 Key study attributes, 98–101
Herb Contraindications & Drug Knowledge, sources of, 2–3
Interactions, 156
Herbal Medicines, 157
Historical control trials, 18 L
Level of significance, 39
I Levels of evidence, 104, 112,
117
Inappropriate test, data ana- control group vs. no control
lyzed, 33–35 group, 93
Inclusion/exclusion criteria, 18, interventional vs. observa-
70–71 tional study design, 92
Individual patient decision, power set/met, 98
108–9, 111, 128–29, 131– prospective vs. retrospec-
38 tive observational study
Inductive reasoning, 3 design, 95
Intent-to-treat analysis, 21, 64 randomized or non-
International Pharmaceutical randomized study design,
Abstracts, 52 97
Internet, as resource, 57–58 study design characteris-
Interpretation basics, 7–8, 11– tics, 88–90
38 summary of study design
Interval data, 27, 28, 31, 33, 35 characteristics, 99
Index 195
LexiComp, 52 Herb & Supplement Refer-
Lexi-Comp Pediatric Dosage ence, 155
Handbook, 67, 68, 69 Nominal data, 27, 28, 31, 33
Likert-type scale, 27, 33, 34 Non-binomial data, 27
Logical reasoning, 2–3, 108 Non-normal data distribution,
Look backward, 94–95 29
Look forward, 94 Non-parametric tests, 29–31,
35
Non-systematic reviews, 122,
M 123
Magnitude of difference, 39 Normal data distribution, 29,
31
Major limitations, 105–7
Number needed to treat, 17
Medical College of Virginia
Hospitals, Drug Informa-
tion Service, 41
Medline, 52, 160
O
Observational study, 91–92, 93
MeSH, 161, 162
look forward, backward,
Meta-analyses, 119–22, 123
94–95
Micromedex, 52
Observational trial design, 14–
Multiple item checklists, 61– 17
62, 84–85
Ordinal data, 27, 28, 31, 33
Outcome measures, 19
N dietary supplements, 165
N of 1 trial, 13–14
Narrative reviews, 122, 123 P
Natural Medicine Comprehen-
Parallel interventional trial, 12,
sive Database, 154, 155,
13
158
Parametric tests, 29–31, 35
Natural Products Association,
167 Per protocol analysis, 21, 79
Natural Standard, 154–55 PICO (Patient, Intervention,
Comparison, Outcome),
Herb & Supplement Hand-
42–45
book, 156, 158
applied illustration, 46
196 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
approach, 48 Prospective design, 95
approach defined, 44–45 observational trial, 14, 15,
illustration, 44 94
Placebo control trials, 17–18 PubMed, 52
Population–based decision, P-value, 19–20, 21, 35
108–9, 110, 111, 129–30 significance and, 38
class review, 138–41
comparative review, 141–
49
Q
Power, 21–26 Quality of evidence, 87, 107–8
components of, 39 equation, 88
dietary supplement studies, Quality seal programs, 167
163
met and no difference
exists, 21–22, 24
R
not met, 65 Randomization, 18, 75–79, 96–
97, 101
not met and difference
exists, 25–26 Ratio data, 27, 28, 31, 33, 35
not met and no difference Recommendation
exists, 25 components required for,
sample size and, 22–23 107
scenarios, 26, 66 confidence of, 110, 112–13
set and met, 24–25, 63–66, decision type and, 109–10
97–98 firm vs. cautious, 127, 128
Practice guidelines, 122–25 format, 113
Product from conclusion, 107–8
dosage, dietary supplement justification, 114–17
studies, 163–64 Recommendation statement,
quality, 166, 167, 168 110
Professional’s Handbook of example, 114
Complementary & Alterna- justification format, 113
tive Medicines, 156, 158
strength of, 112–13
Prospective cohort groups, 94
Relative risk, 16
reduction, 16, 17
Index 197
Retrieve pertinent information, Special considerations/popula-
5–6, 51 tions, 114–15, 116, 129,
dietary supplements, 152, 130
153–62 Statistical significance, 19–21
individual decision, 132– p–value and, 38
33 Statistical tests, 29–35, 78, 79–
Internet as resource, 57–58 80
organizing search results, decision flowchart, 32
55–56 Study design
population-based decision, interventional trial, 12–14
139, 142–43
observational trial, 14–17
search strategy, 53–54
Study design characteristics,
starting decision, 52 88–90
Retrospective design, 95 five questions, 91–98
observational trial, 14–15, summary of, 99
94–95
Study length appropriate, 69
Risk
Summary, conclusion develop-
interpretation, 17 ment, 105–7
ratios, 16–17 Summary Table of Evidence
Evaluated, 104–5
S Systematic reviews, 119–22,
123
Sackett, David, 1, 43
Safety, 114, 116, 129, 130
Sample size, 22, 39
T
dietary supplement studies, Ten Major Considerations, 62–
163 63, 88
reporting, 23 blinding, 74–75
Scientific method, 1, 2, 3 conclusions supported by
results, 82
Search
dosage/treatment appropri-
and sift technique, 55–57,
ate, 67–69
59
evaluation measurements,
organizing results, 55–57
80–81
strategy, 53–54
exclusion criteria, 71–74
terms, 47–48
198 The Pharmacist’s Guide to Evidence-Based Medicine for Clinical Decision Making
inclusion criteria, 70–71
key study attributes, 98–
101
power set/met, 63–66
randomization, 75–79
statistics tests, 79–80
study length appropriate,
69
Third-party laboratory ratings,
167
Traditions, 2
Trial and error, 2
Trial design concepts, 17–19
Type II error rate, 39
U–V
University of Missouri–Kansas
City School of Pharmacy
Drug Information Center, 5
W–Z
Wikipedia, 57